4,510
Views
16
CrossRef citations to date
0
Altmetric
Review Article

Predictors of outcome in childhood Plasmodium falciparum malaria

, &
Pages 199-221 | Received 30 Sep 2019, Accepted 20 Jan 2020, Published online: 17 Feb 2020

ABSTRACT

Plasmodium falciparum malaria is classified as either uncomplicated or severe, determining clinical management and providing a framework for understanding pathogenesis. Severe malaria in children is defined by the presence of one or more features associated with adverse outcome, but there is wide variation in the predictive value of these features. Here we review the evidence for the usefulness of these features, alone and in combination, to predict death and other adverse outcomes, and we consider the role that molecular biomarkers may play in augmenting this prediction. We also examine whether a more personalized approach to predicting outcome for specific presenting syndromes of severe malaria, particularly cerebral malaria, has the potential to be more accurate. We note a general need for better external validation in studies of outcome predictors and for the demonstration that predictors can be used to guide clinical management in a way that improves survival and long-term health.

Introduction

Malaria is a mosquito-transmitted infectious disease caused by protozoan parasites of the genus Plasmodium. Over 150 Plasmodium species have been identified [Citation1] and found to infect a diverse range of hosts from birds and reptiles to deer and monkeys. Infections in humans primarily involve five Plasmodium species: Plasmodium falciparum, P. vivax, P. ovale, P. malariae, and P. knowlesi, although several other species have been observed to cause zoonotic infections.

According to the World Health Organization (WHO), there were 219 million reported cases of malaria across 87 countries in 2017. Countries within the WHO Africa region had the highest incidence and accounted for approximately 92% of malaria cases and 93% of malaria deaths. This corresponds to an estimated 435,000 deaths directly caused by malaria in the same year, the majority (61%) being in children under five [Citation2]. This review will specifically focus on predictors of death and other adverse outcomes in children with malaria.

Many factors can influence the clinical outcomes of malaria, including parasite species, host, and parasite genetics, innate and acquired immunity, access to appropriate treatment, comorbidities, and antimalarial resistance. P. falciparum accounts for the vast majority of severe malaria (SM) and malaria deaths globally, and is the focus of this review, but P. vivax and P. knowlesi can also cause severe and fatal infections, whilst P. malariae and P. ovale almost always cause uncomplicated malaria (UM) [Citation3,Citation4]. In the case of P. falciparum, naturally acquired immunity develops with repeated infections in high transmission settings resulting in the sequential acquisition of protection from severe disease and then from symptomatic infection [Citation5]. In these regions, young children who have not yet acquired immunity are at greatest risk of life-threatening malaria, whereas all age groups typically remain susceptible in the lowest transmission settings where exposure is insufficient to induce immunity [Citation6].

The life cycle of P. falciparum involves two hosts, the female Anopheles mosquito and the human. Unlike the human host, the mosquito suffers no deleterious effects of the infection. In humans, the infection starts when the sporozoite form of the parasite enters the skin during mosquito feeding and then rapidly transits through the bloodstream to the liver where there is a pre-symptomatic incubation period of replication within hepatocytes [Citation7]. After about 1 week, the parasites leave the liver and enter the bloodstream where they invade red blood cells and undergo repeated 48-h cycles of asexual replication [Citation5]. These asexual blood-stage parasites are the cause of symptoms that define the onset of malaria [Citation8]. Some parasites will also differentiate into sexual stage gametocytes, ready for uptake by another mosquito bite and continuation of the transmission cycle, but gametocytes themselves do not cause any clinical symptoms [Citation7].

Like most other infectious diseases, the consequences of infection with P. falciparum can vary widely between individuals, and the observed severity is also influenced by how soon after infection an individual is assessed and treated. Outcomes of infection can range from asymptomatic carriage of parasites (seen in individuals with substantial naturally acquired immunity) to unpleasant influenza-like symptoms, to significant organ dysfunction and death [Citation9]. In this review, we focus only on symptomatic infections. UM is defined by the presence of asexual blood-stage parasites and typical clinical symptoms of malaria such as fever, headache, nausea, and vomiting in the absence of another cause and without the features which define SM [Citation9]. SM is defined by similar criteria except that one or more clinical or laboratory features associated with increased risk of death is also present ().

Table 1. Clinical and laboratory features of severe malaria and their prognostic value

For many laboratory and clinical parameters, the distinction between SM and UM is determined by a threshold value and it is important to note that these thresholds are somewhat artificial for the convenience of classification. For example, hemoglobin (Hb), hematocrit, blood glucose, and lactate levels are all continuous variables that are likely to have a continuous relationship with the risk of death rather than a step-effect at a particular threshold value. It is also important to recognize that SM is a multisystem, multi-organ disease, although SM in children is often categorized into three easily recognized clinical syndromes that can occur separately or in combination: cerebral malaria (CM), severe malarial anemia (SMA), and respiratory distress [Citation10,Citation11].

A substantial amount of work has been conducted to understand what distinguishes the pathogenesis of different SM phenotypes from UM and from each other with the hope of identifying targets for adjunctive therapy. Unfortunately, to date, no adjunctive treatments have proven effective in clinical trials [Citation12]. Similar comparisons have also indicated molecular biomarkers that distinguish between SM and UM. Whilst this in itself is not necessarily useful when SM is already classified based on clinical features and routine laboratory tests, it is reasonable to expect that some biomarkers associated with the occurrence of SM might also predict the risk of death or long-term morbidity.

Prognostic markers for mortality and long-term morbidity could have several applications. Foremost, they could help to determine the most appropriate course of action for each individual patient by informing the choice of medical treatment and whether transfer to a medical facility with higher-level care facilities is required. Secondarily they may be used as inclusion criteria, or for stratification, in clinical trials of adjunctive treatments or interventions to improve outcomes in SM, potentially increasing power to detect effects on the outcome. Third, identification of children with greatest risk of long-term sequelae could potentially be used to target interventions to reduce the impact of these sequelae on their life, even if the sequelae cannot be prevented.

In this literature review, we critically evaluate the usefulness of clinical and laboratory features of SM as predictors of mortality and long-term complications in childhood P. falciparum malaria and assess the potential of molecular biomarkers and other approaches to improve the accuracy of prediction. We consider these questions for SM in general and for the major syndromes of CM, SMA, and respiratory distress individually. We briefly consider the important role of coinfections (human immunodeficiency virus (HIV) and bacteremia) as well as malaria in returning travelers. Finally, we synthesize the existing evidence and propose strategies to improve outcome prediction in pediatric P. falciparum malaria.

Publications considered in this review were identified through keyword searches of the PubMed database using terms specific to each sub-section of the review. Additional publications were identified from our own bibliographies and from references in articles identified through Pubmed. We did not exclude any studies based on study size, and thus studies ranged in size from tens to thousands of patients. The largest study, from the Severe Malaria in African Children (SMAC) network included data from 20,333 patients collected between 2000 and 2003 from 5 sites (Banjul, Blantyre, Kilifi, Kumasi, and Lambarene) [Citation13]. Wherever possible we report confidence intervals for estimates of risk or association, which indicate the precision of these estimates, in part determined by the study size.

Prognostic value of criteria defining severe malaria

The criteria commonly used to define SM were selected because they are associated with adverse outcome, however, they are not all equally good predictors () and the evidence for each will be discussed in more detail below.

One important general caveat is that interpretation of the relationship of prognostic indicators with outcomes is often complicated by the difficulty of knowing whether malaria is the primary or only cause of these features. In some high transmission settings, it is very common for parasitemia to be an incidental finding in children with other febrile illnesses with similar presentations. For example, a child with decreased conscious level due to bacterial meningitis who has incidental parasitemia might be classified as having CM unless suitable diagnostic facilities are available to identify the meningitis. In this case, the prognosis would be more dependent on appropriate treatment of the meningitis than of the malaria parasites.

Another caveat is that direct comparison of the Relative Risk (RR), Hazard Ratio (HR), Odds Ratio (OR), and Adjusted Odds Ratio (AOR) for death between studies requires some caution. Variation in study size and design, different study settings, different populations, and variation in the antimalarial and supportive care treatments available, likely contribute to the heterogeneity of the risk estimates. For example, the increasing use of intravenous artesunate over the last decade has reduced the likelihood of death in SM compared with studies conducted in the pre-artesunate era. Data originating from clinical trials may show lower overall and relative risks of mortality than in observational studies, an effect often attributed to improved clinical care provided during clinical trials. This is particularly evident in the “Artesunate versus quinine in the treatment of severe falciparum malaria in African children trial (AQUAMAT)” by von Seidlein et al. [Citation14], which showed consistently lower risks of death associated with severity features than other studies examining the same clinical features.

Hyperparasitaemia

In many infectious diseases, pathogen load is thought to be linked to outcome [Citation15]. In malaria, high parasitemia can indicate a high parasite replication rate, insufficient clearance of parasites by the host response, and or a longer duration of infection [Citation16]. Peripheral hyperparasitaemia () is considered an important indicator of SM [Citation17,Citation237]. The definition of hyperparasitaemia is dependent on the setting: in low prevalence settings and returning travelers the threshold defining SM is often set at 4–5% of red blood cells [Citation18,Citation19], whereas in areas of stable endemicity the WHO recommends >10% [Citation17,Citation20].

Hyperparasitaemia has been associated with an increased likelihood of symptomatic malaria infections [Citation21] as well as malaria-associated mortality [Citation22]. In multivariable analyses of the SMAC cohort, hyperparasitaemia was associated with mortality (OR of 3.6 [95% CI 1.8–7.5]) in Banjul (The Gambia), but not at the other four study sites [Citation22], and other studies have also found that its prognostic significance diminished when it was combined with other prognostic markers [Citation14,Citation23]. Interestingly, in a study of Kenyan children admitted to hospital over a 27-y period during which the incidence of SM declined dramatically, hyperparasitaemia (defined as parasite density >250,000 parasites per μl) only emerged as a predictor of death during the lowest incidence period [Citation23]. This suggests that the effect of hyperparasitaemia on outcome may only be apparent at lower transmission intensities. It is also important to note that circulating parasitemia is a poor indicator of total body parasite load in P. falciparum malaria because the late asexual blood stage parasites sequester in the microcirculation and are not usually sampled in the blood used to estimate parasitemia [Citation24]. For this reason, it is not surprising that hyperparasitaemia is a relatively poor predictor of outcome, as illustrated in by the low RR values and low prognostic value given by the WHO. Indeed, Silamut & White showed that when these late blood-stage parasites are visible in the circulation it is a grave prognostic marker [Citation25].

Hypoglycemia

Hypoglycemia () has been reported in 1–32% of children presenting to hospital with malaria [Citation26Citation30], with a greater prevalence in children below 3 y of age, those with convulsions or coma, hyperparasitaemia, and those receiving quinine treatment [Citation31]. The pathophysiology is multifactorial and increased glucose utilization, impaired gluconeogenesis, and hyperinsulinemia secondary to quinine therapy all contribute [Citation27,Citation32Citation34]. Many studies have demonstrated that hypoglycemia is an important risk factor for death ()[Citation22,Citation26,Citation27,Citation35,Citation36,Citation237].

Metabolic acidosis

Metabolic acidosis () is a common finding (43–46% prevalence) in studies of African children admitted to hospital with malaria [Citation31,Citation37,Citation38]. It is thought to occur due to a combination of microvascular obstruction by sequestered parasites, vascular endothelial dysfunction, anemia, and inflammation driving increased glycolysis. Metabolic acidosis is strongly associated with an increased risk of death ()[Citation16,Citation35,Citation36,Citation39]. In the AQUAMAT trial, the severity of acidosis was associated with increasing mortality (pH 7.2 associated with almost 20% mortality; pH <7.0 associated with >60% mortality [Citation14]). Hyperlactatemia is a major contributor to the acidaemic state in malaria, along with reduced plasma bicarbonate. Prognosis has been shown to be worse in patients whose lactate levels do not normalize within a few hours of admission and the levels of lactate in fatal cases were nearly double those of survivors (7.1 mmol/L in fatal vs. 3.6 mmol/L in survivors) amongst Gambian children [Citation40].

Respiratory distress

Respiratory distress () describes abnormal, deep and effortful breathing (chest-indrawing or nasal flaring) with an increased respiratory rate for age. The incidence of respiratory distress in SM ranges from 7-41% [Citation10,Citation31,Citation41] and it is a major risk factor for death, but with highly variable estimates of effect size ().It is present in over half of reported malarial deaths [Citation10,Citation14,Citation22,Citation36,Citation42]. In childhood malaria, respiratory distress usually reflects underlying metabolic acidosis while pulmonary edema () and associated acute respiratory distress syndrome are rare in children but more commonly seen in adults [Citation43]. Respiratory distress and metabolic acidosis as a clinical syndrome and markers of prognosis are considered in further detail below.

Renal failure

Renal dysfunction associated with malaria is due to acute tubular necrosis and less commonly interstitial nephritis and glomerulonephritis; multiple mechanisms including parasitized erythrocytes causing tissue hypoperfusion and immune complex deposition have been proposed [Citation44,Citation45]. The optimal assessment of renal function requires serial biochemical measurements of blood and/or urine using a patient’s baseline renal function as a reference [Citation46]. Unfortunately, this is not routinely available in most malaria endemic settings. As diagnostic facilities have improved in some settings, it has been recognized that the burden of acute renal impairment in malaria is substantial [Citation45,Citation47]. A recent Ugandan study assessing the renal function of children with SMA and CM reported a prevalence of over 35% [Citation48]. Results of many clinical studies investigating SM have supported the prognostic significance of renal dysfunction (). Additionally, von Seildlein et al. showed a statistically significant, but small, rise in mortality with increased blood urea nitrogen in a multivariable predictive model (blood urea nitrogen >20 mg/dL AOR 1.02 [95% CI 1.02–1.03])[Citation14]. Moreover, the presence of acute renal impairment has not only been shown to be a risk factor for the development of chronic kidney disease (OR 2.81 [95% CI 1.02–7.73]) but also for long-term neurological impairment in CM (AOR 3.03 [95% CI 1.22–7.58]) at 2-y follow-up [Citation48].

Thrombocytopenia

Thrombocytopenia is defined as platelet levels lower than expected for age. While it is a well-documented finding in SM [Citation49], it rarely causes symptoms. Several underlying mechanisms have been postulated including immune-mediated destruction, splenic sequestration, and bone marrow suppression [Citation49]. Literature on the prognostic value of thrombocytopenia in malaria is contradictory and relatively limited (). Several studies have reported its usefulness in aiding malaria diagnosis (platelets <150, sensitivity 60%, specificity 88% [Citation50]) and as a predictor of mortality () but have varying results due to their different definitions of thrombocytopenia. Furthermore, other studies found that while platelet levels were lower in SM compared to UM patients, this did not have any association with outcome [Citation51,Citation52].

Bleeding

Overt bleeding associated with malaria is uncommon and can be secondary to multiple pathological processes including coagulopathy and thrombocytopenia [Citation53], which can occasionally manifest as bleeding from the mucous membranes and venepuncture sites [Citation17]. When symptoms are present, it is indicative of a more complicated disease process and hence could be speculated to be associated with greater mortality. However, due to its rarity, no studies have formally assessed its value as a marker of prognosis () and the majority of the literature is from case series and reports of adult malaria [Citation54Citation57].

Severe malaria anemia

SMA is the most common complication of P. falciparum infections, affecting the youngest children in areas of high transmission [Citation58Citation62], manifesting clinically with pallor, lethargy, and poor feeding. The WHO define SMA as hemoglobin <5 g/dl or a hematocrit of <15% in children (aged <12 y) with a parasite count >10,000/microliter [Citation17]. SMA is a major cause of hospitalization [Citation60,Citation63] and is associated with poor prognosis and increased mortality, although estimates of effect size vary () [Citation22]. Substantial work has been carried out to understand the pathophysiology of SMA and its value as a prognostic indicator which will be discussed in further detail below.

Prostration

Prostration is a sign of weakness and may also include mild cerebral impairment, resulting in the inability to sit unassisted or to breastfeed in those under the age of 6 months [Citation14,Citation17]. It can progress to a coma or be present in the post-ictal phase following a seizure. While prostration is commonly reported in SM [Citation10,Citation14,Citation64], it can be a subjective observation [Citation17]. This is likely one of the reasons why it has variable prognostic value in studies in African children () [Citation65] ranging from small to moderate increase in the risk of death, and usually considerably less increase in risk than in children with other features of SM () [Citation14,Citation22,Citation36]. This is consistent with the WHO assessment that prostration has a low prognostic value ().

Neurological abnormalities

The neurological abnormalities in SM include seizures and impaired consciousness (). The severity of impaired consciousness is often measured using the Blantyre coma scale. This is a modified version of the Pediatric Glasgow Coma Scale, specifically constructed to allow rapid and straightforward assessment of the severity of malaria-induced coma. Impaired consciousness and seizures are associated with a higher risk of mortality in pediatric malaria patients ()[Citation16,Citation22,Citation36,Citation37,Citation66]. It is important to note that the definition of coma varies between studies and that some studies do not state a formal definition (). This has resulted in some variability in the effect size estimation of impaired consciousness (). The clinical syndrome CM, which encompasses these neurological symptoms is considered in further detail below.

Jaundice

Jaundice is yellowing of the skin, mucosal surfaces, and whites of eyes caused by excess bilirubin, which can be detected by clinical examination. As a feature of SM, it is defined as raised plasma bilirubin (>50 µM or 3 mg/dl) with evidence of significant parasitemia (>100,000 µl) [Citation17]. It is caused by a combination of hemolysis and hepatic dysfunction and is more common in adults, though it can also be a presenting feature in neonatal malaria [Citation17,Citation66]. Several studies have identified the presence of jaundice as a risk factor for mortality in children with malaria in univariable analysis (). In one large study, the mortality in cases with jaundice was 22/114 (19%), whilst without jaundice 505/5312 (10%) (p < 0.001) [Citation14]. However, multivariable analyses have not consistently identified jaundice as an independent predictor of mortality when combined with other features of SM (RR 2.6 [95% CI 1.1–6.3] [Citation10]; OR 0.87 [95% CI 0.31–2.49] [Citation23]), possibly because it is relatively uncommon compared to some of the other features of SM.

Shock

Shock () is a life-threatening condition caused by circulatory failure leading to tissue hypoxia. If recognized and treated in its early stages (compensated shock), it can be reversible, however, it can progress quickly to multiorgan failure and death [Citation67]. In recent years, two large multicentre studies have analyzed the risk of death following shock in malaria. von Seidlein et al. showed a significantly increased mortality rate of 19% in children with shock vs. 9% in those without, in a cohort of children with SM [Citation14]. In another study, the presence of compensated shock was associated with an increased risk of death () on univariable analysis but not in the multivariable analysis due to the effect of shock on mortality rate being accounted for by the other covariates included into the models [Citation23]. illustrates the fairly consistent moderate estimates of the effect of this feature on mortality in univariable analysis. This is reflected by the WHO giving this feature a high prognostic value.

Independence of prognostic markers

Due to the complex, interlinked pathophysiology, many of the features of SM occur together and are correlated. Researchers have tried to identify features with the greatest independent prognostic value using multivariable models.

The most commonly recognized independent factors include metabolic acidosis [Citation14,Citation37,Citation40,Citation68Citation71], hypoglycemia [Citation10,Citation29,Citation40,Citation68Citation70], impaired consciousness [Citation10,Citation14,Citation29,Citation64,Citation68Citation71], renal dysfunction [Citation14,Citation29,Citation69] and respiratory distress [Citation10,Citation29,Citation64,Citation69Citation71]. Those less frequently described in the literature include: the presence of an underlying chronic illness [Citation14], SMA [Citation68,Citation71] and parasite stage visible by microscopy [Citation69]. Combinations of these scores allowed for the prediction of the majority of malaria deaths, for example 92% of the deaths in a cohort of 1844 children (average age: 26.4 months) with a mortality rate of 3.5% were predicted by a model incorporating four features: respiratory distress (RR 3.9 [95% CI 2.0–7.7]), impaired consciousness (RR 3.3 [95% CI 1.6–7.0]), hypoglycemia (RR 3.3 [95% CI 1.6–6.7]), and jaundice (RR 2.6 [95% CI 1.1–6.3])[Citation10]. von Seidlein et al. also combined several clinical scores into a multivariable model of 13 clinical features, which had an area under the curve (AUC) of 0.85 [95% CI 83–87] [Citation14].

Recent metanalyses have also helped to elucidate the strength of multiple prognostic indicators in predicting mortality in SM. Sypniewska et al. evaluated 36 variables from 30 studies of children in Africa with SM; variables most strongly associated with death included renal failure (OR 5.96 [95% CI 2.93–12.11]), coma (OR 4.83 [95% CI 3.11–7.5]), hypoglycemia (OR 4.59 [95% CI 2.68–7.89]), shock (OR 4.31 [95% CI 2.15–8.64]), and deep breathing (OR 3.8 [95% CI 3.29–4.39]) [Citation72]. Leopold et al. applied a causal inference model to a pooled data set of adults and children with SM from 15 countries across Asia and Africa. They reported similar findings of increased mortality associated with acidosis (OR 2.10 for a 7 mEq/L increase in base deficit [95% CI 1.93–2.28]), renal impairment (OR 1.71 for a 2-fold increase in blood urea nitrogen [95% CI 1.58–1.86]), coma (OR 3.59 [95% CI 3.07–4.21]), seizures (OR 1.40 [95% CI 1.16–1.68]), shock (OR 1.51 [95% CI 1.14–1.99]), and presumed pulmonary edema (OR 1.58 [95% CI 1.04–2.39]) [Citation73].

Score systems have been developed to simplify the application of prediction models for rapid implementation in clinical settings. For instance, Helbok et al. developed the Lambaréné Organ Dysfunction Score (LODS) 0–3, which denote the presence of coma, prostration, and deep breathing – the three variables which were most significantly associated with fatal outcomes in six sites across Africa from the SMAC network [Citation64]. The LODS was found to be a simple and useful predictor of mortality (OR for a 1-unit increase in the LODS was 3.53 [95% CI 3.30–3.77])[Citation64]. A second example is Pediatric Early Death Index for Africa (PEDIA) which uses multiple clinical variables with different weighting (jaundice (weight, 1.0); subcostal indrawing (weight, 1.0); prostration ± seizure (weight, 2.0); altered consciousness (with seizures, weight, 2.0; without seizures, weight, 3.0); wasting (weight, 1.0)) [Citation74,Citation75]. PEDIA effectively discriminated survivors and non-survivors in a prospective subgroup analysis performed by Conroy et al. with AUC 0.92 [95% CI 0.90–0.93] [Citation75].

Prognostic biomarkers of severe malaria

In addition to clinical features, many studies have also identified molecular biomarkers associated with adverse outcome in children with malaria. Whilst SM is a heterogeneous condition defined by the variety of clinical manifestations outlined above, biomarkers that predict progression from UM to SM and outcome across all SM phenotypes would be extremely attractive in clinical practice. Potential prognostic biomarkers originating from both host and parasite have been described.

Parasite biomarkers

Plasma levels of P. falciparum Histidine Rich Protein-2 (PfHRP2) have been demonstrated in multiple studies to be associated with the severity of malaria in children [Citation11,Citation76]. PfHRP2 is a parasite protein which is predominantly released from infected blood cells at the time of schizont rupture, and has a relatively long half-life, meaning that levels accumulate in plasma as parasite load increases [Citation77]. PfHRP2 is widely believed to be a better indicator of total body parasite load than parasitemia, because plasma levels of the protein should represent both sequestered and circulating parasites. PfHRP2 concentrations have consistently been found to increase as the severity of infection increases, characterized by the presence of more severe symptoms including coma, acidosis, SMA, and death [Citation11,Citation76,Citation78,Citation79]. For example, in a large study (n = 3,826) Hendriksen et al. found that the mortality of SM patients increased by about 20% for every log10 increase in serum concentration of PfHRP2 above 174 ng/mL [Citation79].

Furthermore, higher plasma concentrations of PfHRP2 are associated with the presence of the CM phenotype (CM: n = 25, UM: n = 125; CM mean PfHRP2 concentration: 7838 ng/mL [SD, 2.5 ng/mL], UM mean PfHRP2 concentration: 421 ng/mL [SD, 9.0 ng/mL], p-value < 0.001 [Citation80]). PfHRP2 is also important because it is one of the antigens detected in many rapid diagnostic tests for P. falciparum [Citation81]. The potential utility of quantitative PfHRP2 tests for prognosis is still being explored. One challenge for this approach is that the concentrations of PfHRP2 associated with SM seem to differ greatly between different transmission settings (in a similar way to the relationship between parasitemia and severity) and so establishing universal thresholds for outcome prediction is unlikely to be possible.

Hemozoin, the by-product of the digestion of hemoglobin by Plasmodium parasites, could be an indirect marker of parasite load. Its presence in parasitized erythrocytes, neutrophils and to a lesser extent monocytes, was associated with increased mortality risk at most sites in the SMAC cohort [Citation36]. However, the C-statistics (equivalent to the AUROC) for these markers ranged from 0.50 (erythrocytes) to 0.70 (granulocytes) [Citation36] indicating relatively low prognostic value. This might be explained by the fact that hemozoin cannot be degraded by phagocytic cells, so its accumulation may be more reflective of cumulative than current parasite load.

Another parasite molecule strongly associated with the outcome of infection is the P. falciparum erythrocyte membrane protein 1 (PfEMP1). PfEMP1 proteins are parasite antigens encoded by var genes and transported onto the surface of parasitized erythrocytes [Citation82Citation84]. They bind cell surface receptors such as CD36 on the vascular endothelium to facilitate the sequestration of parasitized red cells within small blood vessels and avoid transit through the spleen [Citation85]. Only one var gene is expressed by an individual parasite at any time; however, in each new generation, daughter parasites can switch expression amongst the 60+ var genes in their genome. The var genes and the PfEMP1 proteins they encode are highly polymorphic, enabling them to evade host antibody responses [Citation86,Citation87,]. Despite this, it has been possible to classify them into groups with conserved structural and biological properties.

Preferential expression of different subsets of PfEMP1 is associated with different disease manifestations [Citation88Citation93]. Having a significantly higher proportion of var genes that are group A and/or B that contain domains that bind the Endothelial Protein C Receptor (EPCR) has consistently been found to be associated with the SM phenotypes CM and SMA in different populations (mixed adult and child cohorts [Citation91Citation94]). It is thought that PfEMP1 binding to EPCR not only facilitates sequestration and increased parasite load, but also has direct pathogenic effects by impeding the function of EPCR and leading to dysregulation of vascular inflammation, coagulation, and the permeability of the vascular endothelium [Citation95]. Indeed, a recent study has shown that the combination of var expression profile and parasite load is a better predictor of clinical phenotype (SM vs. UM) than parasite load alone [Citation96], however, the study was not designed to detect association with survival. Although the determination of var gene or PfEMP1 expression might have useful prognostic value, applying this to routine clinical practice would currently be challenging due to the extensive genetic variability and complexity of classification of var genes.

Parasite molecules may be associated with prognosis, independent of any role in pathogenesis, if they influence the success of antimalarial treatment. Numerous genetic variants causing or associated with resistance to different antimalarials have been identified in P. falciparum [Citation97,Citation98], including emerging resistance to current first-line artemisinin-based combination therapies [Citation99]. Available evidence indicates that being infected with parasites resistant to the empirical antimalarial treatment is likely to result in an increased risk of adverse outcome, although data quantifying this risk for specific combinations of resistance mutations and antimalarial drugs is not widely available [Citation97,Citation100]. Nevertheless, rapid identification of resistance mutations at the point-of-care could help to ensure that individuals receive optimal treatment.

Host biomarkers

An appropriate host inflammatory and immune response to infection is vital to help clear parasites from the circulation, whilst an inappropriate or excessive response could result in damage to host tissues. Thus, there has been great interest in the potential of components of the host inflammatory and regulatory responses as predictors of outcome. For example, the prototypical pro-inflammatory cytokine, tumor necrosis factor-α (TNF), was one of the first to be found to be expressed at higher levels in SM than UM, and at highest levels in fatal cases, and this has been confirmed repeatedly since [Citation101Citation103]. These findings led to adjunctive clinical trials of anti-TNF monoclonal antibody treatment in SM, which ultimately showed no improvements in outcome [Citation103].

Interleukin 10 (IL-10) is an anti–inflammatory cytokine also implicated in the dysregulation of inflammation. In mouse models, it has been found to be protective against SM phenotypes, including CM [Citation104Citation106], but has also been found to reduce the ability of the host to clear the parasites from the blood [Citation107]. Studies of African children indicate that IL-10 may prevent the manifestation of some severe phenotypes by suppressing the expression of TNF [Citation108] but in doing so may promote the growth of the parasite, which in itself can cause SM [Citation109,Citation110].

The ratio of different combinations of inflammatory and regulatory cytokines has also been highlighted as important in balancing the inflammatory response. For example, higher ratios of transforming growth factor-beta (TGF-β1)/IL-12 and IL-10/IL-12 have been found in children with SM relative to UM [Citation102].

Differences in the levels of numerous other plasma proteins and small molecules have been observed in comparisons of children with SM with UM, and some of these have been evaluated as prognostic markers. For example, the soluble form of triggering receptor expressed on myeloid cells-1 (sTREM-1), whose function is to amplify the inflammatory response of neutrophils and monocytes, and soluble vascular endothelial growth factor receptor 1 (sFlt-1), which is involved in cell proliferation, has been associated with outcome. With plasma concentration cutoff points of >289.9 pg/mL and >1066.3 pg/mL for sTREM-1 and sFLT-1, respectively, both achieved high sensitivity values of 95.7% [95% CI 78.1–99.9] and 82.6 [95% CI 61.2–95.0], respectively, for predicting mortality in SM patients, but specificity was low (sTREM-1: 43.8% [95% CI 32.7–55.3], sFlt-1: 57.5 [95% CI 45.9–68.5]) [Citation111].

In contrast, the cytokine interferon gamma-induced protein-10 (IP-10), which is involved in the chemoattraction of T cells, natural killer cells, and monocytes, achieved high values for both sensitivity of 82.6% [95% CI 61.2–95.0] and specificity of 85% [95% CI 75.3–92.0] for mortality prediction in this cohort [Citation111] when the plasma concentration was >831.2 pg/mL. Other inflammatory response proteins associated with mortality in SM include high-mobility group protein-1 (HMGB1, a cytokine) [Citation112], monocyte CD36 expression [Citation113] and von Willebrand factor (VWF, a glycoprotein in the blood) [Citation114]. Thus, many proteins associated with inflammation have potential prognostic value in SM, however, many require further validation in different patient cohorts to better assess this potential.

It is likely that the combination of multiple host biomarkers will be needed to produce widely applicable predictive tests for the outcome of malaria. The selection of optimal panels of biomarkers remains to be achieved, although modern variable selection and machine learning approaches should make this easier [Citation115,Citation116]. The incremental benefit of biomarkers above the predictive value of clinical features should be considered, as well as the possibility of prediction rules combining both clinical features and biomarkers to achieve the best possible prediction.

Clinical syndromes and specific prognostic biomarkers

In addition to prognostic features and biomarkers for SM in general, there are predictors of outcome specific to each of the major phenotypes of SM which occur in children: CM, SMA, and respiratory distress. Since there are clear clinical and biological differences between these syndromes [Citation11], a more tailored approach to prognosis may improve prediction for individual patients.

Cerebral malaria

Mortality

CM is one of the most feared clinical complications of malaria. The pathogenesis is still debated, but in P. falciparum malaria the sequestration of parasites in the microvasculature of the brain appears to be a necessary feature, and cerebral edema (brain swelling) appears to be the final common pathway leading to death. Mortality rate is variable depending on the setting and presence of other SM features, however, even when these are accounted for it can still be as high as 15-25% in tertiary care centers [Citation117,Citation118].

CM is diagnosed in a patient with P. falciparum identified on a peripheral blood smear, with one or more neurological symptoms including reduced consciousness, coma (Blantyre coma score ≤ 2) and seizures, where other potential causes have been ruled out. Other causes could include hypoglycemia, hyperpyrexia, acidosis, SMA, medication, post-ictal phase, as well as encephalopathy caused by a co-infection. How to exclude these other causes and thus identify the true-CM patients has been discussed at length [Citation37,Citation119,Citation120].

In 2011, Beare et al. suggested that the presence of malaria-specific retinopathy (changes at the back of the eye which can be seen during clinical examination) should be required for the diagnosis of CM [Citation121]. These features include retinal whitening, hemorrhages, and changes to the vasculature. The vasculature of the retina and brain has a common embryonic origin, and so changes in the retinal vessels are thought to be representative of the sequestration occurring in the brain microvasculature [Citation122]. The severity of retinopathy is also being investigated as an independent predictor of mortality in CM patients [Citation123]. In one study the OR for mortality based on the presence of any signs of retinopathy was 5.5 [95% CI 1.3, 23.4] [Citation123]. Whether retinopathy-negative patients with features of CM and no alternative diagnosis genuinely have CM remains a subject of debate.

A substantial amount of work has been conducted to identify what distinguishes fatal-CM from non-fatal-CM. Sequential studies using increasingly powerful Magnetic Resonance Imaging (MRI) scanners have revealed that increased intracranial pressure due to cerebral edema is the cause of death in African children with CM [Citation118,Citation124Citation126]. Higher-resolution scans have been used to try and understand the etiology of the cerebral edema and have identified obstruction of the cerebral microvasculature, microhaemorrhages, and evidence of autoregulatory dysfunction [Citation127]. A small study of Indian children with CM indicated that impairment of the blood-brain barrier (due to sequestration) was responsible for the swelling in the posterior region of the brain [Citation128]. However, another small study in Taiwanese children with UM also found some similar MRI abnormalities in the absence of CM [Citation129]. The prognostic value of specific MRI features remains to be defined and at present, the potential of MRI for routine risk stratification is unrealistic in most malaria endemic settings due to the cost and logistic challenges of scanning seriously ill children.

Electroencephalography (EEG), which measures the electrical activity of the brain, has also been used to try to identify predictive features for mortality in CM. Large studies of children with CM in Malawi and Uganda found that lower average voltage, lower maximum voltage, and a lack of reactivity were all associated with death [Citation118,Citation130]. However, further research is required to better understand the added value of EEG as a prognostic tool.

Many potential molecular biomarkers have been identified as associated with CM mortality. Biomarkers have been identified at the proteomic level (i.e. 266 host and parasite proteins identified by Moussa et al. [Citation131]) and at the genetic and metabolomic levels (i.e. 19 plasma metabolites identified by Gupta et al. [Citation132]) and originate from both the host and parasite. Those from the host are mainly involved in inflammatory responses (). Many of these biomarkers require prospective validation in larger cohorts of patients before their clinical utility can be accurately assessed.

Table 2. Examples of proteins and genes identified as associated with CM mortality

The vascular growth factors, angiopoietin-1, and angiopoietin-2, involved in angiogenesis and regulation of vascular endothelial function, are two of the most well-established biomarkers for prediction of outcome and there is evidence from experimental CM in mice of the former having a mechanistic role in the development of CM [Citation112]. These proteins are predictors of mortality for both CM specifically as well as the general SM phenotype in a range of settings and populations, with angiopoietin-2 achieving area under the receiver operating characteristics (AUROC) values between 0.71 and 0.83 () [Citation111,Citation133,Citation134]. These proteins are also currently being investigated as therapeutic targets [Citation134].

Neurological sequelae

Survivors of CM are at risk of long-term neurological impairments (neurological sequelae) including deficits in cognitive, motor, and/or sensory function. The most common neurological sequelae following CM in children are muscular weakness, co-ordination/balance, and speech problems [Citation135,Citation136]. The attribution of such broadly defined neurological sequelae to malaria is challenging [Citation137], even with expensive longitudinal studies [Citation136]. However, the best estimates indicate that 5-25% of CM survivors will exhibit some form of neurological sequelae 6 months after discharge depending on how carefully they are assessed [Citation135,Citation138Citation140]. Additionally, around 10% of pediatric CM survivors are thought to develop epilepsy [Citation139,Citation141].

The most well-established predictors of neurological sequelae appear to be the depth and duration of coma [Citation29,Citation136,Citation142] and the presence of convulsions [Citation135,Citation143Citation145]. However, Jallow et al. found that having a Blantyre coma score of equal to or less than 2 predicted the development of neurological sequelae with only modest accuracy (sensitivity: 0.74, specificity: 0.69, AUC: 0.72, [95% CI 0.67–0.76]) [Citation29]. Many of these features also share molecular biomarkers with neurological sequelae [Citation146Citation149], such as kynurenine, which is associated with the duration of coma as well as numerous measures of neurological sequelae including attention deficit [Citation147]. The severity of retinopathy has also been linked to the presence of neurological sequelae, with papilledema, disk hyperemia, macula whitening, and foveal annulus being most strongly associated with neurodevelopmental test outcomes [Citation150].

Neurological sequelae have also been reported in children with SM and UM who did not have CM, when measured relative to healthy controls [Citation151]. For example, Ugandan patients who had SMA had more behavioral problems relative to age matched healthy controls at 12 and 24 months after their index malaria episode [Citation136]. Despite adjusting for potential confounders in the analysis, it is unclear whether this is a direct effect of malaria (i.e. brain damage) or an indirect effect (for example, due to preexisting poor nutrition or reduced learning opportunities).

The relationship between hypoglycemia and neurological sequelae is also uncertain. A case-control study in Nigerian children identified that hypoglycemia was more common in children with neurological sequelae at discharge after CM (7/11 children, 63.6%) than those without neurological sequelae (7/51 children, 13.7% [Citation152]). However, a subsequent larger study of 160 children (13.7% of survivors had neurological sequalae on discharge) in the same setting found no statistically significant association between hypoglycemia and the presence of neurological sequelae [Citation140] and nor did a large study of 624 children (23.3% with neurological sequalae on discharge) conducted in the Gambia [Citation135]. One explanation for these discrepancies may be due to the latter two studies having more representative cohorts of patients with neurological sequelae, or due to differences in the time to recognition and treatment of hypoglycemia between these studies.

Other molecular biomarkers have also been identified as predictors of neurological sequelae in pediatric malaria, most notably, erythropoietin [Citation145]. Erythropoietin, a hormone and cytokine best known for controlling the production of red blood cells, has been identified as a potential protectant against neurological damage in a variety of settings [Citation153]. Plasma levels of erythropoietin greater than 200 units per liter have been found to be associated with an 80% reduction in the risk of long-term neurological impairment in CM [Citation145]. However, evidence from its use in clinical trials to treat other neurological conditions such as cerebral ischemia (against which it has also been shown to be neuroprotective [Citation154]) has suggested that high erythropoietin levels can also result in higher fatality rates [Citation155]. This is consistent with another study which found that high plasma levels of erythropoietin were associated with longer coma in CM and a 1.7-fold increase in mortality [95% CI 1.09–2.8] [Citation156], but only in children with Hb levels <8 g/dL. This further complicates the potential utility of erythropoietin as a prognostic biomarker for CM patients.

Vascular endothelial growth factor (VEGF) was also investigated by Casals-Pascual et al., but found not to be neuroprotective [Citation145]. However, its plasma levels were inversely correlated with erythropoietin and it was associated with the presence of seizures (OR: 4.1 [95% CI 1.75–9.60]) [Citation145].

Proteins and metabolites in the cerebrospinal fluid (CSF) have been identified as potential biomarkers of neurological damage and predictors of neurological sequelae in CM. For example, high CSF (but not plasma levels) [Citation157] of TNF were significantly associated with the occurrence of neurological impairment in one study of Ugandan children, but with a relatively small effect size and only in children older than 5 y [Citation148]. Tau protein levels in CSF were associated with a greater severity of neurological sequelae assessed by overall cognitive scores, working memory, and attention at 2 y after discharge [Citation149]. CSF tau protein levels measured at hospital admission predicted the presence of neurological sequelae at time of discharge relatively poorly (AUC 0.62 [95% CI 0.52–0.72]), but were a better predictor of persistent deficits at 24 months (AUC 0.89 [95% CI 0.8–0.99] [Citation149]), suggesting that this could be a promising biomarker. Elevated CSF kynurenic acid and kynurenine at presentation have also been found to correlate with decreased scores in behavioral tests at 12 months after index CM episode [Citation147,Citation148]. However, all these biomarkers require validation in different pediatric patient cohorts, and many seem to only be associated with specific aspects of cognition in older children (>5 y).

Severe malarial anemia

SMA () is one of the most common complications associated with childhood SM in endemic settings, resulting in a high burden for health services [Citation158]. SMA can arise chronically from repeated or persistent infections or acutely due to severe hemolysis, sometimes associated with blackwater fever. Blackwater fever is a rare complication in children characterized by rapid intravascular hemolysis, fever, and acute renal failure; reported in association with quinine use and glucose-6-phosphate dehydrogenase deficiency [Citation159Citation161] and associated with increased risk of mortality (HR 3.37 [95% CI 1.3–8.5]) and hospital readmission (HR 1.68 [95% CI 1.1–2.5]) [Citation162]. Hemolysis has also been reported to occur 1–2 weeks following treatment with artesunate, primarily in nonimmune travelers, resulting in an increased need for blood transfusions [Citation163].

Multiple factors can contribute to SMA, including co-infections with HIV, helminths (such as hookworm), and bacteria, and nutritional deficiencies [Citation62,Citation164,Citation165]. Furthermore, malaria-driven natural selection for hemoglobinopathies and erythrocyte abnormalities themselves result in increased risk of severe anemia during an acute malarial illness [Citation166Citation170]. Alongside hemolysis of parasitized erythrocytes, there is accelerated destruction of non-parasitized erythrocytes, becoming more prominent with increasing disease severity [Citation158,Citation171Citation174]. Abnormal erythrocytes can be sequestered by the rapidly enlarging spleen, sometimes progressing to massive hyper-reactive splenomegaly [Citation175]. There is additional suppression and dysregulation of erythropoiesis during the acute illness [Citation176Citation178] thought to be triggered by the host immune response and associated with intramedullary deposition of malaria pigment, hemozoin [Citation179Citation181]. The disease process is further exacerbated by slower reticulocyte response in immunologically naive individuals and repeated infections restricting bone marrow recovery [Citation36,Citation158].

Risk of death in SMA is dependent on several factors including the degree of anemia [Citation14,Citation182], rate of disease progression [Citation17,Citation183], age, transmission intensity [Citation184], access to and quality of health services, and availability of prompt transfusions [Citation158]. The relationship between these factors and SMA is consistently stated in the literature but the effect is not well quantified. If SMA is accompanied by another clinical syndrome, particularly respiratory distress, the mortality rates are much higher (SMA alone ~1%; SMA and severe respiratory distress ~16% [Citation10,Citation185]) but outcomes can be improved with timely blood transfusions [Citation42,Citation186].

There is also evidence from clinical studies, epidemiological findings, and mouse-models for proposing a causal relationship between SMA and risk of bacterial co-infection, and hence higher case fatality [Citation187,Citation188]. The most plausible mechanism being hemolysis resulting in favorable conditions for bacterial growth and impaired neutrophil function [Citation188].

Using a causal modeling approach, Leopold et al., showed that moderate anemia (hematocrit 15–25%) was in fact protective against mortality (OR 0.87 for a decrease of 10 percentage points in hematocrit [95% CI 0.80–0.95]) with no evidence for a beneficial effect of transfusion (OR for death in the transfused group of 0.99 [95% CI 0.97–1.02]) [Citation73]. This protective effect was not seen in the retrospective analysis by Maitland et al., where reduction in admission hemoglobin levels was associated with a small increase in odds of mortality (hemoglobin 6–6.9 OR 1.03[95% CI 0.95–1.12]; hemoglobin 5–5.9 OR 1.18 [95% CI 1.08–1.30]; hemoglobin 4–4.9 OR 1.12 [95% CI 0.97–1.29]; hemoglobin 3.9–3 OR 1.33 [95% CI 1.06–1.69]) with greatest mortality risk at hemoglobin levels below 3 g/dl (OR 6.36 [95% CI 4.21–9.62]) [Citation189].

Treatment with blood transfusion is recommended when hemoglobin levels fall below 5g/dl [Citation18]. While lifesaving, especially in settings where readily and safely available [Citation31], transfusions can carry a significant risk of transmitting other bloodborne infections such as HIV [Citation190]. In the past, up to 50% of children admitted with malaria in some hyperendemic settings received blood transfusions [Citation42,Citation191,Citation192]. Retrospective analysis in Kenyan children suggests that pre-transfusion hemoglobin levels may influence the potential benefit of blood transfusion. Perhaps surprisingly, transfusion at a baseline hemoglobin level of 4–4.9 g/dL was associated with increased risk of death (OR of death 3.29 [95% CI 1.21–8.94], compared to un-transfused children), while mortality tended to be lower in transfused children with pre-transfusion hemoglobin less than 3.9 (Hb 3.9–3 g/dL, OR 0.59 [95% CI 0.21–1.69]; Hb < 3g/dL OR 0.72 [95% CI 0.29–1.78]) though these values were not statistically significant, possibly due to lack of statistical power [Citation189].

Many biomarkers associated with the presence of/or susceptibility to SMA have been identified. Interesting findings include a dysregulated immune response, primarily Th1 cytokines, and chemokines, and their association with dyserythropoiesis [Citation193Citation197]. Genetic variants that confer either susceptibility (TNF promoter variants [Citation198]; IL-10 promoter variant [Citation199]) or protection against (SCFC promoter variant [Citation200]) SMA have also been identified. While this work has furthered our understanding of underlying pathophysiological mechanisms, the roles of these biomarkers as prognostic indicators in the clinical setting are limited as clinical symptoms and basic laboratory tests adequately identify the clinical syndrome and its severity. Further work is required to identify biomarkers and signatures of poor prognosis following the development of SMA and indicators of associated long-term complications.

Respiratory distress and metabolic acidosis

In childhood malaria, respiratory distress is largely attributed to metabolic acidosis () and is less commonly due to parenchymal lung disease or heart failure related to anemia [Citation17,Citation201,Citation202]. Respiratory distress due to malaria shares many clinical features with pneumonia, and it is often difficult to distinguish the two when diagnostic facilities are limited [Citation203Citation205].

Over 80% of children with SM complicated by respiratory distress are found to be profoundly acidotic [Citation10]. Moreover, deep breathing can accurately predict the presence of acidosis (sensitivity 91%, specificity 83% [Citation202]). When appropriate tests are available, metabolic acidosis has consistently been identified as an independent predictor of mortality [Citation10,Citation14,Citation37,Citation40,Citation69,Citation206,Citation207]. In a meta-analysis of studies the OR for death was 2.3 [95% CI 1.2–4.4] [Citation72]. Furthermore, high (78%) and early mortality was reported in cases with hyperkalemia in the context of acidosis in SM [Citation208].

The principle cause of acidosis is thought to be tissue hypoxia and resultant anaerobic glycolysis generating lactic acid, and additional accumulation and impairment of the metabolism of lactic acid [Citation40] and ketone bodies [Citation38,Citation209]. There is emerging evidence for the role of other organic acids, including hydroxyphenyllactic acid (HPLA), α-hydroxybutyric acid, and β-hydroxybutyric acid [Citation210] and numerous products of the gut microbiota discovered in a recent metabolomic study [Citation211]. Further research is necessary to define if levels of these more recently identified metabolic acids will improve the prediction of outcome.

Coinfections and prognosis

Bacterial coinfection

Malaria, young age, malnutrition, and HIV infection were identified as the strongest risk factors for bacteremia amongst children admitted to hospital in a large study in Kenya [Citation212]. Children with SM are known to have a higher prevalence of bacteremia; most commonly non-typhoidal Salmonella [Citation212Citation214]. Several pathological processes have been proposed including translocation of gram-negative organisms across the bowel wall, specific macrophage and neutrophil dysfunction, and functional hyposplenism [Citation187 Citation188 Citation215,,]. Bacterial co-infection in malaria patients has been associated with a higher risk of death in some [Citation212 Citation216,] but not all studies [Citation217].

HIV coinfection

The geographical overlap between high malaria incidence and HIV prevalence presents a substantial burden caused by coinfection. Whilst literature on the pediatric population is limited, due to the lower prevalence of HIV in children, studies have shown that HIV-infected individuals of all ages are more susceptible to SM [Citation218Citation224] – especially in areas of unstable endemicity [Citation17,Citation223,Citation225]. Co-infected individuals have increased parasitemia, a more complicated disease course, higher morbidity, and higher mortality. For example, one study [Citation219,Citation226] identified a mortality of 26% (19/74) in HIV-coinfected children versus 9% (53/581) in uninfected children. Increased mortality rates have been observed in HIV co-infected children with CM compared with HIV-uninfected children with CM (OR 1.4 [95% CI, 1.1–1.9][Citation227]). Co-infected children are also at greater risk of severe anemia (OR 8.71 [95% CI 3.37–22.51])[Citation218,Citation228].

Prognosis of malaria in nonimmune travelers

Travelers to malaria endemic areas may be malaria naïve with no naturally acquired immunity or may have waning immunity even if they were previously resident in the endemic country. Most life-threatening malaria complications occur in nonimmune travelers returning from malaria endemic areas, with 5% of those affected suffering SM [Citation229]. The mortality rate in malaria cases in Europe was 0.4%-3% [Citation230Citation232] between 1987 and 2006, predominantly due to P. falciparum malaria. Due to the low prevalence of malaria and low associated mortality in Europe, no study has been performed to identify risk factors for death or poor long-term prognosis specifically in children in the context of best available supportive care. Therefore, definitions of SM and risk factors have been extrapolated from endemic settings. In the UK, surveillance data showed that children, pregnant women, and older people were particularly at risk of SM, but mortality was highest in those over 65 y of age with very few deaths in children [Citation232,Citation233]. Factors associated with more severe disease include high levels of parasitemia, peripheral P. falciparum blood schizonts, pigment deposits in leucocytes, metabolic acidosis, male sex, delay in seeking treatment, diagnosis, or receiving care, nonuse, or inappropriate use of chemoprophylaxis, coma, and renal impairment [Citation232,Citation233].

Conclusion

Severe childhood malaria is a multi-system disease that encompasses a range of clinical complications including CM, SMA, and respiratory distress. The defining features of SM differ considerably in their frequency, the strength of evidence for their association with adverse outcomes, and their predictive value. Many studies have combined multiple features into prediction models and scoring systems, which has improved the accuracy of prediction and increased ease of use. The most consistent and important predictors of death appear to be coma, markers of metabolic derangement (acidosis and hypoglycemia), and renal failure.

Numerous molecular biomarkers have been proposed to predict outcome in SM, including PfHRP2 concentrations, var gene expression profiles, sTREM-1, sFlt-1, and angiopoietins 1 and 2. However, their potential may be compromised by technical challenges to implementation in clinical tests (var genes), difficulty in identifying universal thresholds across different transmission settings (PfHRP2), and incomplete understanding of complex interactions with other host response proteins (cytokines). To overcome the latter, feature selection techniques could be implemented to identify the best panels of non-redundant biomarkers and clinical features that together give more accurate outcome predictions. Few studies have included validation of prediction in external cohorts, but this is essential in order to provide evidence that they may be generalizable.

It is clear that prognostic factors and biomarkers specific for distinct SM syndromes also hold promise, and may result in better prediction of adverse outcomes than general prognostic markers in SM. This parallels ongoing debate about whether syndrome-specific adjunctive treatments for SM are needed, essentially applying the principles of personalized medicine to malaria. This approach to prognosis appears most promising in CM, and additional work is needed to identify and validate syndrome-specific biomarkers in SMA and respiratory distress. Perhaps more importantly, there is a large gap between the identification of prognostic features and biomarkers and providing evidence that their measurement contributes to improved short or long-term outcomes for patients. Identification of predictors of neurological sequelae for CM now needs to be combined with intervention studies to prevent or mitigate the effects of these sequelae.

In the ongoing search for predictors of outcome in malaria, we believe that it is essential to consider how biomarkers and other predictors could be used in malaria endemic settings. A successful predictive test is likely to be cheap, rapid, and easy to measure with minimal requirement for infrastructure or specialized training. It should be applicable across different settings, not confounded by the age or immune status of the patient, and it should be highly accurate. Ideally, the results of the prognostic test would guide realistic treatment choices for each individual patient, benefitting those at most risk of death or long-term morbidity.

In this literature review, we have critically evaluated the usefulness and potential of the clinical and molecular features associated with SM mortality and long-term sequelae. However, there is still a need for a systematic meta-analysis of the impact of all these features on prognosis prediction accounting for the heterogeneity between the associated studies. This heterogeneity includes the differences in the sample sizes, study settings, and age range of the patients studied, making a systematic meta-analysis extremely challenging.

Abbreviations

AOR=

Adjusted Odds Ratio

AUC=

Area Under the Curve

AUROC=

Area Under the Receiver Operating Characteristic

CI=

Confidence Interval

CM=

Cerebral Malaria

CSF=

Cerebrospinal Fluid

EEG=

Electroencephalography

EPCR=

Endothelial Protein C Receptor

Hb=

Haemoglobin

HIV=

Human Immunodeficiency Virus

HMGB1=

High-Mobility Group Protein-1

HPLA=

Hydroxyphenyllactic Acid

HR=

Hazards Ratio

IL=

Interleukin

IP10=

Interferon gamma-induced protein-10

LODS=

Lambaréné Organ Dysfunction Score

MRI=

Magnetic Resonance Imaging

OR=

Odds Ratio

PEDIA=

Paediatric Early Death Index for Africa

PfEMP1=

P. falciparum Erythrocyte Membrane Protein 1

PfHRP2=

P. falciparum Histidine Rich Protein-2

RR=

Relative Risk

sFlt-1=

Soluble Vascular Endothelial Growth Factor Receptor 1

SMA=

Severe Malarial Anaemia

SMAC=

Severe Malaria in African Children

sTREM-1=

Soluble form of Triggering Receptor Expressed on Myeloid cells-1

TGF-β=

Transforming Growth Factor beta

TNF=

Tumour Necrosis Factor

UM=

Uncomplicated Malaria

VEGF=

Vascular Endothelial Growth Factor

VWF=

Von Willebrand factor

WHO=

World Health Organisation

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

This work was supported by an Engineering and Physical Sciences Research Council studentship (awarded to C.D.), the Medical Research Council (MRC) and the UK Department for International Development (DFID) under the MRC/DFID Concordat agreement that is also part of the EDCTP2 program supported by the European Union [MR/L006529/1 to A.J.C.] and infrastructure support from National Institute for Health Research Imperial Biomedical Research Centre.

References

  • Ramasamy R. Zoonotic malaria – global overview and research and policy needs. Front Public Health. 2014 Aug 18;2:123.
  • World Health Organisation. World Malaria Report 2018. Geneva: World Health Organization; 2018.
  • Mendis K, Sina BJ, Marchesini P, et al. The neglected burden of Plasmodium vivax malaria. Am J Trop Med Hyg. 2001 Feb;64(1–2 Suppl):97–106.
  • Cox-Singh J, Davis TME, Lee K-S, et al. Plasmodium knowlesi malaria in humans is widely distributed and potentially life threatening. Clin Infect Dis. 2008 Jan 15;46(2):165–171.
  • White NJ, Pukrittayakamee S, Hien TT, et al. Malaria. Lancet. 2014 Feb 22;383(9918):723–735.
  • Snow RW, Marsh K. The consequences of reducing transmission of Plasmodium falciparum in Africa. Adv Parasitol. 2002;52:235–264.
  • Phillips MA, Burrows JN, Manyando C, et al. Malaria. Nat Rev Dis Primer. 2017 Aug;3(3):17050.
  • Tuteja R. Malaria − an overview. FEBS J. 2007;274(18):4670–4679.
  • World Health Organisation. WHO Malaria Terminology.Geneva: World Health Organization; 2019.
  • Marsh K, Forster D, Waruiru C, et al. Indicators of life-threatening malaria in African children. N Engl J Med. 1995 May 25;332(21):1399–1404.
  • Cunnington AJ, Walther M, Riley EM. Piecing together the puzzle of severe malaria. Sci Transl Med. 2013 Nov 13;5(211):211ps18.
  • Varo R, Crowley VM, Sitoe A, et al. Adjunctive therapy for severe malaria: a review and critical appraisal. Malar J. 2018 Jan 24;17(1):47.
  • Taylor T, Olola C, Valim C, et al. Standardized data collection for multi-center clinical studies of severe malaria in African children: establishing the SMAC network. Trans R Soc Trop Med Hyg. 2006 Jul;100(7):615–622.
  • von Seidlein L, Olaosebikan R, Hendriksen ICE, et al. Predicting the clinical outcome of severe falciparum malaria in African children: findings from a large randomized trial. Clin Infect Dis. 2012 Apr;54(8):1080–1090.
  • Cunnington AJ. The importance of pathogen load. PLoS Pathog. 2015 Jan 8;11(1):e1004563.
  • Georgiadou A, Lee HJ, Walther M, et al. Modelling pathogen load dynamics to elucidate mechanistic determinants of host– Plasmodium falciparum interactions. Nat Microbiol. 2019 Sep;4(9):1592–1602.
  • World Health Organization. Severe Malaria. Trop Med Int Health. 2014 Sep;19(1):7–131.
  • World Health Organisation. Guidelines for the Treatment of Malaria. 3rd ed. Geneva: World Health Organization; 2015.
  • Centers for Disease Control and Prevention - malaria - about malaria - disease [Internet]. 2019 [cited 2019 Sep 28]. Available from: https://www.cdc.gov/malaria/about/disease.html
  • Lalloo DG, Shingadia D, Bell DJ, et al. UK malaria treatment guidelines 2016. J Infect. 2016 Jun 1;72(6):635–649.
  • Smith T, Schellenberg JA, Hayes R. Attributable fraction estimates and case definitions for malaria in endemic areas. Stat Med. 1994 Nov 30;13(22):2345–2358.
  • Kendjo E, Agbenyega T, Bojang K, et al. Mortality patterns and site heterogeneity of severe malaria in African children. PLoS One. 2013 Mar 7;8(3):e58686.
  • Njuguna P, Maitland K, Nyaguara A, et al. Observational study: 27 years of severe malaria surveillance in Kilifi, Kenya. BMC Med. 2019 Jul 8;17(1):124.
  • Cunnington AJ, Riley EM, Walther M. Stuck in a rut? Reconsidering the role of parasite sequestration in severe malaria syndromes. Trends Parasitol. 2013 Dec;29(12):585–592.
  • Silamut K, White NJ. Relation of the stage of parasite development in the peripheral blood to prognosis in severe falciparum malaria. Trans R Soc Trop Med Hyg. 1993 Jul 1;87(4):436–443.
  • Taylor TE, Molyneux ME, Wirima JJ, et al. Blood glucose levels in Malawian children before and during the administration of intravenous quinine for severe falciparum malaria. N Engl J Med. 1988 Oct 20;319(16):1040–1047.
  • White NJ. The pharmacokinetics of quinine and quinidine in malaria. Acta Leiden. 1987;55:65–76.
  • Osier FHA, Berkley JA, Ross A, et al. Abnormal blood glucose concentrations on admission to a rural Kenyan district hospital: prevalence and outcome. Arch Dis Child. 2003 Jul 1;88(7):621–625.
  • Jallow M, Casals-Pascual C, Ackerman H, et al. Clinical features of severe malaria associated with death: a 13-year observational study in The Gambia. PLoS One. 2012 Sep 28;7(9):e45645.
  • Bassat Q, Guinovart C, Sigaúque B, et al. Malaria in rural Mozambique Part II: Children Admitted to Hospital. Malar J. 2008 Feb 26;7(1):37.
  • Dondorp AM, Fanello CI, Hendriksen IC, et al. Artesunate versus quinine in the treatment of severe falciparum malaria in African children (AQUAMAT): an open-label, randomised trial. Lancet. 2010 Nov 13;376(9753):1647–1657.
  • White NJ, Warrell DA, Chanthavanich P, et al. Severe hypoglycemia and hyperinsulinemia in falciparum malaria. N Engl J Med. 1983 Jul 14;309(2):61–66.
  • Thien HV, Kager PA, Sauerwein HP. Hypoglycemia in falciparum malaria: is fasting an unrecognized and insufficiently emphasized risk factor? Trends Parasitol. 2006 Sep 1;22(9):410–415.
  • Madrid L, Lanaspa M, Maculuve S, et al. Malaria-associated hypoglycaemia in children. Expert Rev Anti Infect Ther. 2014 Dec;26(13):1–11.
  • Camara B, Diagne NR, Faye PM, et al. Critères de gravité et facteurs pronostiques du paludisme chez l’enfant à Dakar. Médecine Mal Infect. 2011 Feb 1;41(2):63–67.
  • Kremsner PG, Valim C, Missinou MA, et al. Prognostic value of circulating pigmented cells in African children with malaria. J Infect Dis. 2009 Jan 1;199(1):142–150.
  • Taylor TE, Borgstein A, Molyneux ME. Acid-base status in paediatric Plasmodium falciparum malaria. Q J Med. 1993 Feb;86(2):99–109.
  • English M, Sauerwein R, Waruiru C, et al. Acidosis in severe childhood malaria. QJM. 1997 Apr 1;90(4):263–270.
  • Newton CRJC, Valim C, Krishna S, et al. The prognostic value of measures of acid/base balance in pediatric falciparum malaria, compared with other clinical and laboratory parameters. Clin Infect Dis. 2005 Oct 1;41(7):948–957.
  • Krishna S, Waller DW, Kuile Ter F, et al. Lactic acidosis and hypoglycaemia in children with severe malaria: pathophysiological and prognostic significance. Trans R Soc Trop Med Hyg. 1994 Jan 1;88(1):67–73.
  • Genton B, D’Acremont V, Rare L, et al. Plasmodium vivax and mixed infections are associated with severe malaria in children: a prospective cohort study from Papua New Guinea. PLoS Med. 2008 Jun 17;5(6):e127.
  • Lackritz EM, Campbell CC, Ruebush TK, et al. Effect of blood transfusion on survival among children in a Kenyan hospital. Lancet. 1992 Aug 29;340(8818):524–528.
  • Van den Steen PE, Deroost K, Deckers J, et al. Pathogenesis of malaria-associated acute respiratory distress syndrome. Trends Parasitol. 2013 Jul;29(7):346–358.
  • Silva da GB, Pinto JR, Barros EJG, et al. Kidney involvement in malaria: an update. Rev Inst Med Trop Sao Paulo. 2017;59:e53.
  • Elsheikha HM, Sheashaa HA. Epidemiology, pathophysiology, management and outcome of renal dysfunction associated with Plasmodia infection. Parasitol Res. 2007 Jul 13;101(5):1183.
  • Inker AL, Perrone R. Assessment of kidney function [Internet]. UpToDate. [cited 2019 Oct 8]. Available from: https://www.uptodate.com/contents/assessment-of-kidney-function
  • Conroy AL, Hawkes M, Elphinstone RE, et al. Acute kidney injury is common in pediatric severe malaria and is associated with increased mortality. Open Forum Infect Dis. 2016 Apr 1;3(2):ofw046.
  • Conroy AL, Opoka RO, Bangirana P, et al. Acute kidney injury is associated with impaired cognition and chronic kidney disease in a prospective cohort of children with severe malaria. BMC Med. 2019 May 21;17(1):98.
  • Lacerda MVG, Mourão MPG, Coelho HCC, et al. Thrombocytopenia in malaria: who cares? Mem Inst Oswaldo Cruz. 2011 Aug;106:52–63.
  • Lathia TB, Joshi R. Can hematological parameters discriminate malaria from nonmalarious acute febrile illness in the tropics? Indian J Med Sci. 2004 Jun;58(6):239–244.
  • Ladhani S, Lowe B, Cole AO, et al. Changes in white blood cells and platelets in children with falciparum malaria: relationship to disease outcome. Br J Haematol. 2002;119(3):839–847.
  • Chimalizeni Y, Kawaza K, Taylor T, et al. The platelet count in cerebral malaria, is it useful to the clinician? Am J Trop Med Hyg. 2010 Jul;83(1):48–50.
  • Francischetti IMB, Seydel KB, Monteiro RQ. Blood coagulation, inflammation, and malaria. Microcirculation. 2008;15(2):81–107.
  • Kochar DK, Das A, Kochar A, et al. Thrombocytopenia in Plasmodium falciparum, Plasmodium vivax and mixed infection malaria: a study from Bikaner (Northwestern India). Platelets. 2010;21(8):623–627.
  • George P, Hegde N. Haematemesis: an uncommon presenting symptom of Plasmodium falciparum malaria. J Clin Diagn Res. 2013 May;7(5):917–918.
  • Wiwanitkit V. Overt bleeding in malarial patients: experience and review. Blood Coagul Fibrinolysis. 2008 Jan;19(1):1–4.
  • Kochhar R, Goenka MK, Mehta S, et al. Gastrointestinal bleeding in malaria. Indian J Gastroenterol. 1990 Oct;9(4):295–296.
  • Snow RW, de Azevedo BI, Lowe BS, et al. Severe childhood malaria in two areas of markedly different falciparum transmission in East Africa. Acta Trop. 1994 Sep 1;57(4):289–300.
  • Snow RW, Guerra CA, Noor AM, et al. The global distribution of clinical episodes of Plasmodium falciparum malaria. Nature. 2005 Mar;434(7030):214–217.
  • Slutsker L, Taylor TE, Wirima JJ, et al. In-hospital morbidity and mortality due to malaria-associated severe anaemia in two areas of Malawi with different patterns of malaria infection. Trans R Soc Trop Med Hyg. 1994 Sep 1;88(5):548–551.
  • Modiano D, Sirima BS, Sawadogo A, et al. Severe malaria in Burkina Faso: influence of age and transmission level on clinical presentation. Am J Trop Med Hyg. 1998 Oct;59(4):539–542.
  • Calis JCJ, Phiri KS, Faragher EB, et al. Severe anemia in Malawian children. N Engl J Med. 2008 Feb 28;358(9):888–899.
  • Biemba G, Dolmans D, Thuma PE, et al. Severe anaemia in Zambian children with Plasmodium falciparum malaria. Trop Med Int Health TM IH. 2000 Jan;5(1):9–16.
  • Helbok R, Kendjo E, Issifou S, et al. The Lambaréné Organ Dysfunction Score (LODS) is a simple clinical predictor of fatal malaria in African children. J Infect Dis. 2009 Dec 15;200(12):1834–1841.
  • Mabeza GF, Moyo VM, Thuma PE, et al. Predictors of severity of illness on presentation in children with cerebral malaria. Ann Trop Med Parasitol. 1995 Jun;89(3):221–228.
  • Anand AC, Puri P. Jaundice in malaria. J Gastroenterol Hepatol. 2005;20(9):1322–1332.
  • Gaieski FD, Mikkelsen EM. Definition, classification, etiology, and pathophysiology of shock in adults [Internet]. UpToDate. [cited 2019 Oct 9]. Available from: https://www.uptodate.com/contents/definition-classification-etiology-and-pathophysiology-of-shock-in-adults?search=definition-classification–etiology-and-pathophysiology-of-shock-in-adults2&source=search_result&selectedTitle=1~150&usage_type=default&display_rank=1
  • Maitland K, Levin M, English M, et al. Severe P. falciparum malaria in Kenyan children: evidence for hypovolaemia. QJM. 2003 Jun 1;96(6):427–434.
  • Waller D, Krishna S, Crawley J, et al. Clinical features and outcome of severe malaria in Gambian children. Clin Infect Dis. 1995 Sep 1;21(3):577–587.
  • Dzeing-Ella A, Nze Obiang PC, Tchoua R, et al. Severe falciparum malaria in Gabonese children: clinical and laboratory features. Malar J. 2005 Jan;9(4):1.
  • Evans JA, May J, Ansong D, et al. Capillary refill time as an independent prognostic indicator in severe and complicated malaria. J Pediatr. 2006 Nov 1;149(5):676–681.
  • Sypniewska P, Duda JF, Locatelli I, et al. Clinical and laboratory predictors of death in African children with features of severe malaria: a systematic review and meta-analysis. BMC Med. 2017 Aug 3;15(1):147.
  • Leopold SJ, Watson JA, Jeeyapant A, et al. Investigating causal pathways in severe falciparum malaria: A pooled retrospective analysis of clinical studies. PLoS Med. 2019 Aug;16(8):e1002858.
  • Berkley JA, Ross A, Mwangi I, et al. Prognostic indicators of early and late death in children admitted to district hospital in Kenya: cohort study. BMJ. 2003 Feb 15;326(7385):361.
  • Conroy AL, Hawkes M, Hayford K, et al.Prospective validation of pediatric disease severity scores to predict mortality in Ugandan children presenting with malaria and non-malaria febrile illness. Crit Care. 2015 Feb 23;19(1):47.
  • Hendriksen ICE, White LJ, Veenemans J, et al. Defining falciparum-malaria-attributable severe febrile illness in moderate-to-high transmission settings on the basis of plasma PfHRP2 concentration. J Infect Dis. 2013 Jan 15;207(2):351–361.
  • Dondorp AM, Desakorn V, Pongtavornpinyo W, et al. Estimation of the total parasite biomass in acute falciparum malaria from plasma PfHRP2. PLoS Med. 2005 Aug 23;2(8):e204.
  • Rubach MP, Mukemba J, Florence S, et al. Plasma Plasmodium falciparum histidine-rich protein-2 concentrations are associated with malaria severity and mortality in Tanzanian children. PLoS One. 2012 May 7;7(5):e35985.
  • Hendriksen ICE, Mwanga-Amumpaire J, von Seidlein L, et al. Diagnosing severe falciparum malaria in parasitaemic African children: a prospective evaluation of plasma PfHRP2 measurement. PLoS Med. 2012;9(8):e1001297.
  • Fox LL, Taylor TE, Pensulo P, et al. Histidine-rich protein 2 plasma levels predict progression to cerebral malaria in Malawian children with Plasmodium falciparum infection. J Infect Dis. 2013 Aug 1;208(3):500–503.
  • Viana GMR, Okoth SA, Silva-Flannery L, et al. Histidine-rich protein 2 (pfhrp2) and pfhrp3 gene deletions in Plasmodium falciparum isolates from select sites in Brazil and Bolivia. PLoS One. 2017 Mar 16;12(3):e0171150.
  • Aley SB, Sherwood JA, Howard RJ. Knob-positive and knob-negative Plasmodium falciparum differ in expression of a strain-specific malarial antigen on the surface of infected erythrocytes. J Exp Med. 1984 Nov 1;160(5):1585–1590.
  • Deitsch KW, Dzikowski R. Variant gene expression and antigenic variation by malaria parasites. Annu Rev Microbiol. 2017 Sep 8;71:625–641.
  • Wahlgren M, Goel S, Akhouri RR. Variant surface antigens of Plasmodium falciparum and their roles in severe malaria. Nat Rev Microbiol. 2017 Aug;15(8):479–491.
  • Turner L, Lavstsen T, Berger SS, et al. Severe malaria is associated with parasite binding to endothelial protein C receptor. Nature. 2013 Jun 27;498(7455):502–505.
  • Scherf A, Hernandez-Rivas R, Buffet P, et al. Antigenic variation in malaria: in situ switching, relaxed and mutually exclusive transcription of var genes during intra-erythrocytic development in Plasmodium falciparum. EMBO J. 1998 Sep 15;17(18):5418–5426.
  • Smith JD. The role of PfEMP1 adhesion domain classification in Plasmodium falciparum pathogenesis research. Mol Biochem Parasitol. 2014 Jul;195(2):82–87.
  • Bull PC, Lowe BS, Kortok M, et al. Parasite antigens on the infected red cell surface are targets for naturally acquired immunity to malaria. Nat Med. 1998 Mar;4(3):358–360.
  • Nielsen MA, Staalsoe T, Kurtzhals JAL, et al. Plasmodium falciparum variant surface antigen expression varies between isolates causing severe and nonsevere malaria and is modified by acquired immunity. J Immunol. 2002 Apr 1;168(7):3444–3450.
  • Rottmann M, Lavstsen T, Mugasa JP, et al. Differential expression of var gene groups is associated with morbidity caused by Plasmodium falciparum infection in Tanzanian children. Infect Immun. 2006 Jul;74(7):3904–3911.
  • Falk N, Kaestli M, Qi W, et al. Analysis of Plasmodium falciparum var genes expressed in children from Papua New Guinea. J Infect Dis. 2009 Jul;1(200):347–356.
  • Warimwe GM, Keane TM, Fegan G, et al. Plasmodium falciparum var gene expression is modified by host immunity. Proc Natl Acad Sci. 2009 Dec 22;106(51):21801–21806.
  • Jespersen JS, Wang CW, Mkumbaye SI, et al. Plasmodium falciparum var genes expressed in children with severe malaria encode CIDRα1 domains. EMBO Mol Med. 2016 Aug 1;8(8):839–850.
  • Jensen ATR, Magistrado P, Sharp S, et al. Plasmodium falciparum associated with severe childhood malaria preferentially expresses PfEMP1 encoded by group A var genes. J Exp Med. 2004 May 3;199(9):1179–1190.
  • Bouwens EAM, Stavenuiter F, Mosnier LO. Cell painting with an engineered EPCR to augment the protein C system. Thromb Haemost. 2015 Nov 25;114(6):1144–1155.
  • Duffy F, Bernabeu M, Babar PH, et al. Meta-analysis of plasmodium falciparum var signatures contributing to severe malaria in African children and Indian adults. mBio. 2019 30;10(2):e00217–19.
  • Blasco B, Leroy D, Fidock DA. Antimalarial drug resistance: linking Plasmodium falciparum parasite biology to the clinic. Nat Med. 2017 Aug 4;23(8):917–928.
  • Haldar K, Bhattacharjee S, Safeukui I. Drug resistance in Plasmodium. Nat Rev Microbiol. 2018 Mar;16(3):156–170.
  • Woodrow CJ, White NJ. The clinical impact of artemisinin resistance in Southeast Asia and the potential for future spread. FEMS Microbiol Rev. 2017;41(1):34–48.
  • Lubell Y, Dondorp A, Guérin PJ, et al. Artemisinin resistance – modelling the potential human and economic costs. Malar J. 2014 Nov 23;13(1):452.
  • Grau GE, Taylor TE, Molyneux ME, et al. Tumor necrosis factor and disease severity in children with falciparum malaria. N Engl J Med. 1989 Jun 15;320(24):1586–1591.
  • Perkins DJ, Weinberg JB, Kremsner PG. Reduced interleukin-12 and transforming growth factor-beta1 in severe childhood malaria: relationship of cytokine balance with disease severity. J Infect Dis. 2000 Sep;182(3):988–992.
  • van Hensbroek MB, Palmer A, Onyiorah E, et al. The effect of a monoclonal antibody to tumor necrosis factor on survival from childhood cerebral malaria. J Infect Dis. 1996 Nov;174(5):1091–1097.
  • Kossodo S, Monso C, Juillard P, et al. Interleukin-10 modulates susceptibility in experimental cerebral malaria. Immunology. 1997 Aug;91(4):536–540.
  • Sanni LA, Jarra W, Li C, et al. Cerebral edema and cerebral hemorrhages in interleukin-10-deficient mice infected with plasmodium chabaudi. Infect Immun. 2004 May 1;72(5):3054–3058.
  • Do Rosario FAP, Langhorne J. T cell-derived IL-10 and its impact on the regulation of host responses during malaria. Int J Parasitol. 2012 May 15;42(6):549–555.
  • Weidanz WP, Batchelder JM, Flaherty P, et al. Plasmodium chabaudi adami: use of the B-cell-deficient mouse to define possible mechanisms modulating parasitemia of chronic malaria. Exp Parasitol. 2005 Oct;111(2):97–104.
  • Othoro C, Lal AA, Nahlen B, et al. A low interleukin-10 tumor necrosis factor-alpha ratio is associated with malaria anemia in children residing in a holoendemic malaria region in western Kenya. J Infect Dis. 1999 Jan;179(1):279–282.
  • Zhang G, Manaca MN, McNamara-Smith M, et al. Interleukin-10 (IL-10) polymorphisms are associated with IL-10 production and clinical malaria in young children. Infect Immun. 2012 Jul 1;80(7):2316–2322.
  • Kumar R, Ng S, Engwerda C. The role of IL-10 in malaria: a double edged sword. Front Immunol. 2019 Feb 12;10:229.
  • Erdman LK, Dhabangi A, Musoke C, et al. Combinations of host biomarkers predict mortality among Ugandan children with severe malaria: a retrospective case-control study. PLoS One. 2011 Feb 25;6(2):e17440.
  • Higgins SJ, Xing K, Kim H, et al. Systemic release of high mobility group box 1 (HMGB1) protein is associated with severe and fatal Plasmodium falciparum malaria. Malar J. 2013 Mar 19;12:105.
  • Cserti-Gazdewich CM, Dhabangi A, Musoke C, et al. Cytoadherence in paediatric malaria: ABO blood group, CD36, and ICAM1 expression and severe Plasmodium falciparum infection. Br J Haematol. 2012 Oct;159(2):223–236.
  • Graham SM, Chen J, Chung DW, et al. Endothelial activation, haemostasis and thrombosis biomarkers in Ugandan children with severe malaria participating in a clinical trial. Malar J. 2016 Feb 2;15(1):56.
  • Herberg JA, Kaforou M, Wright VJ, et al. Diagnostic test accuracy of a 2-transcript host RNA signature for discriminating bacterial vs viral infection in febrile children. JAMA. 2016 Aug 23;316(8):835–845.
  • Wright VJ, Herberg JA, Kaforou M, et al. Diagnosis of Kawasaki disease using a minimal whole-blood gene expression signature. JAMA Pediatr. 2018 01;172(10):e182293.
  • Birbeck GL, Molyneux ME, Kaplan PW, et al. Blantyre Malaria Project Epilepsy Study (BMPES) of neurological outcomes in retinopathy-positive paediatric cerebral malaria survivors: a prospective cohort study. Lancet Neurol. 2010 Dec;9(12):1173–1181.
  • Seydel KB, Kampondeni SD, Valim C, et al. Brain swelling and death in children with cerebral malaria. N Engl J Med. 2015 Mar 19;372(12):1126–1137.
  • Bejon P, Berkley JA, Mwangi T, et al. Defining childhood severe falciparum malaria for intervention studies. PLoS Med. 2007 Aug;4(8):e251.
  • Maitland K. Management of severe paediatric malaria in resource-limited settings. BMC Med. 2015 Mar 3;13(1):42.
  • Beare NAV, Lewallen S, Taylor TE, et al. Redefining cerebral malaria by including malaria retinopathy. Future Microbiol. 2011 Mar;6(3):349–355.
  • MacCormick IJC, Beare NAV, Taylor TE, et al. Cerebral malaria in children: using the retina to study the brain. Brain. 2014 Aug;137(8):2119–2142.
  • Singh J, Verma R, Tiwari A, et al. Retinopathy as a prognostic marker in cerebral malaria. Indian Pediatr. 2016 Apr 1;53(4):315–317.
  • Potchen MJ, Kampondeni SD, Seydel KB, et al. Acute brain MRI findings in 120 Malawian children with cerebral malaria: new insights into an ancient disease. Am J Neuroradiol. 2012 Oct;33(9):1740–1746.
  • Mohanty S, Taylor TE, Kampondeni S, et al. Magnetic resonance imaging during life: the key to unlock cerebral malaria pathogenesis? Malar J. 2014 Jul;18(13):276.
  • Taylor TE, Molyneux ME. The pathogenesis of pediatric cerebral malaria: eye exams, autopsies and neuro-imaging. Ann N Y Acad Sci. 2015 Apr;1342(1):44–52.
  • Potchen MJ, Kampondeni SD, Seydel KB, et al. 1.5 tesla magnetic resonance imaging to investigate potential etiologies of brain swelling in pediatric cerebral malaria. Am J Trop Med Hyg. 2018;98(2):497–504.
  • Mohanty S, Benjamin LA, Majhi M, et al. Magnetic resonance imaging of cerebral malaria patients reveals distinct pathogenetic processes in different parts of the brain. mSphere. 2017 June 7;2(3):e00193–17.
  • Laothamatas J, Sammet CL, Golay X, et al. Transient lesion in the splenium of the corpus callosum in acute uncomplicated falciparum malaria. Am J Trop Med Hyg. 2014 Jun;90(6):1117–1123.
  • Postels DG, Wu X, Li C, et al. Admission EEG findings in diverse paediatric cerebral malaria populations predict outcomes. Malar J. 2018 May 22;17(1):208.
  • Moussa EM, Huang H, Thézénas ML, et al. Proteomic profiling of the plasma of Gambian children with cerebral malaria. Malar J. 2018 Sep 24;17(1):337.
  • Gupta S, Seydel K, Miranda-Roman MA, et al. Extensive alterations of blood metabolites in pediatric cerebral malaria. PLoS One. 2017;12(4):e0175686.
  • Conroy AL, Hawkes M, McDonald CR, et al. Host biomarkers are associated with response to therapy and long-term mortality in pediatric severe malaria. Open Forum Infect Dis. 2016 Sep;3(3):ofw134.
  • de Jong GM, Slager JJ, Verbon A, et al. Systematic review of the role of angiopoietin-1 and angiopoietin-2 in Plasmodium species infections: biomarkers or therapeutic targets? Malar J. 2016 Dec 1;15(1):581.
  • van Hensbroek MB, Palmer A, Jaffar S, et al. Residual neurologic sequelae after childhood cerebral malaria. J Pediatr. 1997 Jul 1;131(1):125–129.
  • Ssenkusu JM, Hodges JS, Opoka RO, et al. Long-term behavioral problems in children with severe malaria. Pediatrics. 2016 Nov;138(5):e20161965.
  • Berkley JA, Mwangi I, Mellington F, et al. Cerebral malaria versus bacterial meningitis in children with impaired consciousness. QJM. 1999 Mar;92(3):151–157.
  • Kihara M, Carter JA, Newton CRJC. The effect of Plasmodium falciparum on cognition: a systematic review. Trop Med Int Health. 2006 Apr;11(4):386–397.
  • John CC, Bangirana P, Byarugaba J, et al. Cerebral malaria in children is associated with long-term cognitive impairment. Pediatrics. 2008 Jul;122(1):e92–99.
  • Oluwayemi IO, Brown BJ, Oyedeji OA, et al. Neurological sequelae in survivors of cerebral malaria. Pan Afr Med J. 2013 Jul 6;15:88.
  • Idro R, Marsh K, John CC, et al. Cerebral malaria: mechanisms of brain injury and strategies for improved neurocognitive outcome. Pediatr Res. 2010 Oct;68(4):267–274.
  • Boivin MJ. Effects of early cerebral malaria on cognitive ability in Senegalese children. J Dev Behav Pediatr. 2002 Oct;23(5):353–364.
  • Meremikwu MM, Asindi AA, Ezedinachi E. The pattern of neurological sequelae of childhood cerebral malaria among survivors in Calabar, Nigeria. Cent Afr J Med. 1997 Aug;43(8):231–234.
  • Idro R, Jenkins NE, Newton CRJC. Pathogenesis, clinical features, and neurological outcome of cerebral malaria. Lancet Neurol. 2005 Dec;4(12):827–840.
  • Casals-Pascual C, Idro R, Gicheru N, et al. High levels of erythropoietin are associated with protection against neurological sequelae in African children with cerebral malaria. Proc Natl Acad Sci U S A. 2008 Feb 19;105(7):2634–2639.
  • Medana IM, Lindert R-B, Wurster U, et al. Cerebrospinal fluid levels of markers of brain parenchymal damage in Vietnamese adults with severe malaria. Trans R Soc Trop Med Hyg. 2005 Aug;99(8):610–617.
  • Holmberg D, Franzén-Röhl E, Idro R, et al. Cerebrospinal fluid kynurenine and kynurenic acid concentrations are associated with coma duration and long-term neurocognitive impairment in Ugandan children with cerebral malaria. Malar J. 2017 28;16(1):303.
  • Shabani E, Ouma BJ, Idro R, et al. Elevated cerebrospinal fluid tumour necrosis factor is associated with acute and long-term neurocognitive impairment in cerebral malaria. Parasite Immunol. 2017 Jul;39(7):e12438.
  • Datta D, Conroy AL, Castelluccio PF, et al. Elevated cerebrospinal fluid tau protein concentrations on admission are associated with long-term neurologic and cognitive impairment in Ugandan children with cerebral malaria. Clin Infect Dis. 2019.
  • Boivin MJ, Vokhiwa M, Sikorskii A, et al. Cerebral malaria retinopathy predictors of persisting neurocognitive outcomes in Malawian children. Pediatr Infect Dis J. 2014 Aug;33(8):821–824.
  • Bangirana P, Musisi S, Boivin MJ, et al. Malaria with neurological involvement in Ugandan children: effect on cognitive ability, academic achievement and behaviour. Malar J. 2011 Nov;3(10):334.
  • Bondi FS. The incidence and outcome of neurological abnormalities in childhood cerebral malaria: a long-term follow-up of 62 survivors. Trans R Soc Trop Med Hyg. 1992 Feb;86(1):17–19.
  • Marti HH, Bernaudin M, Petit E, et al. Neuroprotection and angiogenesis: dual role of erythropoietin in brain ischemia. News Physiol Sci. 2000 Oct;15(5):225–229.
  • Sirén AL, Fratelli M, Brines M, et al. Erythropoietin prevents neuronal apoptosis after cerebral ischemia and metabolic stress. Proc Natl Acad Sci U S A. 2001 Mar 27;98(7):4044–4049.
  • Ehrenreich H, Weissenborn K, Prange H, et al. Recombinant human erythropoietin in the treatment of acute ischemic stroke. Stroke. 2009 Dec;40(12):e647–656.
  • Shabani E, Opoka RO, Idro R, et al. High plasma erythropoietin levels are associated with prolonged coma duration and increased mortality in children with cerebral malaria. Clin Infect Dis. 2015 Jan 1;60(1):27–35.
  • John CC, Panoskaltsis-Mortari A, Opoka RO, et al. Cerebrospinal fluid cytokine levels and cognitive impairment in cerebral malaria. Am J Trop Med Hyg. 2008 Feb;78(2):198–205.
  • White NJ. Anaemia and malaria. Malar J. 2018 Oct 19;17(1):371.
  • Tran TH, Day NP, Ly VC, et al. Blackwater fever in southern Vietnam: a prospective descriptive study of 50 cases. Clin Infect Dis. 1996 Dec;23(6):1274–1281.
  • Bodi JM, Nsibu CN, Longenge RL, et al. Blackwater fever in Congolese children: a report of clinical, laboratory features and risk factors. Malar J. 2013 Jun 15;12(1):205.
  • Recht J, Ashley E, White N. Safety of 8-aminoquinoline antimalarial medicines. Geneva: World Health Organization; 2014.
  • Opoka RO, Waiswa A, Harriet N, et al. Blackwater fever in Ugandan children with severe anemia is associated with poor postdischarge outcomes: a prospective cohort study. Clin Infect Dis. 2019.
  • Jauréguiberry S, Thellier M, Ndour PA, et al. Delayed-onset hemolytic anemia in patients with travel-associated severe malaria treated with artesunate, France, 2011–2013. Emerg Infect Dis. 2015 May;21(5):804–812.
  • Crawley J. Reducing the burden of anemia in infants and young children in malaria-endemic countries of Africa: from evidence to action. Am J Trop Med Hyg. 2004 Aug;71(2 Suppl):25–34.
  • Ghosh K, Ghosh K. Pathogenesis of anemia in malaria: a concise review. Parasitol Res. 2007 Nov;101(6):1463–1469.
  • Weatherall DJ. Genetic variation and susceptibility to infection: the red cell and malaria. Br J Haematol. 2008 May;141(3):276–286.
  • Casanova J-L. Human genetic basis of interindividual variability in the course of infection. Proc Natl Acad Sci U S A. 2015 Dec 22;112(51):E7118–7127.
  • Uyoga S, Ndila CM, Macharia AW, et al. Glucose-6-phosphate dehydrogenase deficiency and the risk of malaria and other diseases in children in Kenya: a case-control and a cohort study. Lancet Haematol. 2015 Oct;2(10):e437–444.
  • Williams TN. Sickle cell disease in Sub-Saharan Africa. Hematol Oncol Clin North Am. 2016 Apr;30(2):343–358.
  • May J, Evans JA, Timmann C, et al. Hemoglobin variants and disease manifestations in severe falciparum malaria. JAMA. 2007 May 23;297(20):2220–2226.
  • Looareesuwan S, Merry AH, Phillips RE, et al. Reduced erythrocyte survival following clearance of malarial parasitaemia in Thai patients. Br J Haematol. 1987 Dec;67(4):473–478.
  • Looareesuwan S, Davis TM, Pukrittayakamee S, et al. Erythrocyte survival in severe falciparum malaria. Acta Trop. 1991 Feb;48(4):263–270.
  • Jakeman GN, Saul A, Hogarth WL, et al. Anaemia of acute malaria infections in non-immune patients primarily results from destruction of uninfected erythrocytes. Parasitology. 1999 Aug;119(Pt 2):127–133.
  • Buffet PA, Safeukui I, Deplaine G, et al. The pathogenesis of Plasmodium falciparum malaria in humans: insights from splenic physiology. Blood. 2011 Jan 13;117(2):381–392.
  • Leoni S, Buonfrate D, Angheben A, et al. The hyper-reactive malarial splenomegaly: a systematic review of the literature. Malar J. 2015 Apr;29(14):185.
  • Phillips RE, Looareesuwan S, Warrell DA, et al. The importance of anaemia in cerebral and uncomplicated falciparum malaria: role of complications, dyserythropoiesis and iron sequestration. Q J Med. 1986 Mar;58(227):305–323.
  • Pathak VA, Ghosh K. Erythropoiesis in malaria infections and factors modifying the erythropoietic response. Anemia. 2016;2016:9310905.
  • Wickramasinghe SN, Abdalla SH. Blood and bone marrow changes in malaria. Baillieres Best Pract Res Clin Haematol. 2000 Jun;13(2):277–299.
  • Awandare GA, Ouma Y, Ouma C, et al. Role of monocyte-acquired hemozoin in suppression of macrophage migration inhibitory factor in children with severe malarial anemia. Infect Immun. 2007 Jan;75(1):201–210.
  • Casals-Pascual C, Kai O, Cheung JOP, et al. Suppression of erythropoiesis in malarial anemia is associated with hemozoin in vitro and in vivo. Blood. 2006 Oct 15;108(8):2569–2577.
  • Aguilar R, Moraleda C, Achtman AH, et al. Severity of anaemia is associated with bone marrow haemozoin in children exposed to Plasmodium falciparum. Br J Haematol. 2014 Mar;164(6):877–887.
  • Kiguli S, Maitland K, George EC, et al. Anaemia and blood transfusion in African children presenting to hospital with severe febrile illness. BMC Med. 2015 Feb 2;13:21.
  • Bartoloni A, Zammarchi L. Clinical aspects of uncomplicated and severe malaria.Mediterr J Hematol Infect Dis. 2012 May 4;4(1):e2012026.
  • Douglas NM, Lampah DA, Kenangalem E, et al. Major burden of severe anemia from non-falciparum malaria species in Southern Papua: a hospital-based surveillance study. PLoS Med. 2013 Dec;10(12):e1001575; discussion e1001575.
  • Bojang KA, Van Hensbroek MB, Palmer A, et al. Predictors of mortality in Gambian children with severe malaria anaemia. Ann Trop Paediatr. 1997 Dec;17(4):355–359.
  • English M, Ahmed M, Ngando C, et al. Blood transfusion for severe anaemia in children in a Kenyan hospital. Lancet. 2002 Feb 9;359(9305):494–495.
  • Takem EN, Roca A, Cunnington A. The association between malaria and non-typhoid Salmonella bacteraemia in children in sub-Saharan Africa: a literature review. Malar J. 2014 Oct;13(13):400.
  • Orf K, Cunnington AJ. Infection-related hemolysis and susceptibility to Gram-negative bacterial co-infection. Front Microbiol. 2015 Jun 30;6:666.
  • Maitland K, Ohuma EO, Mpoya A, et al. Informing thresholds for paediatric transfusion in Africa: the need for a trial. Wellcome Open Res. 2019 Aug;12(4):27.
  • Ekvall H. Malaria and anemia. Curr Opin Hematol. 2003 Mar;10(2):108–114.
  • Obonyo CO, Steyerberg EW, Oloo AJ, et al. Blood transfusions for severe malaria-related anemia in Africa: a decision analysis. Am J Trop Med Hyg. 1998 Nov;59(5):808–812.
  • Colebunders R, Ryder R, Francis H, et al. Seroconversion rate, mortality, and clinical manifestations associated with the receipt of a human immunodeficiency virus-infected blood transfusion in Kinshasa, Zaire. J Infect Dis. 1991 Sep;164(3):450–456.
  • Haldar K, Mohandas N. Malaria, erythrocytic infection, and anemia. Hematol Am Soc Hematol Educ Program. 2009;2009:87–93.
  • Were T, Hittner JB, Ouma C, et al. Suppression of RANTES in children with Plasmodium falciparum malaria. Haematologica. 2006 Oct;91(10):1396–1399.
  • Kurtzhals JA, Adabayeri V, Goka BQ, et al. Low plasma concentrations of interleukin 10 in severe malarial anaemia compared with cerebral and uncomplicated malaria. Lancet. 1998 Jun 13;351(9118):1768–1772.
  • McDevitt MA, Xie J, Ganapathy-Kanniappan S, et al. A critical role for the host mediator macrophage migration inhibitory factor in the pathogenesis of malarial anemia. J Exp Med. 2006 May 15;203(5):1185–1196.
  • Keller CC, Yamo O, Ouma C, et al. Acquisition of hemozoin by monocytes down-regulates interleukin-12 p40 (IL-12p40) transcripts and circulating IL-12p70 through an IL-10-dependent mechanism: in vivo and in vitro findings in severe malarial anemia. Infect Immun. 2006 Sep;74(9):5249–5260.
  • McGuire W, Hill AV, Allsopp CE, et al. Variation in the TNF-alpha promoter region associated with susceptibility to cerebral malaria. Nature. 1994 Oct 6;371(6497):508–511.
  • Ouma C, Davenport GC, Were T, et al. Haplotypes of IL-10 promoter variants are associated with susceptibility to severe malarial anemia and functional changes in IL-10 production. Hum Genet. 2008 Dec;124(5):515–524.
  • Ouma C, Keller CC, Davenport GC, et al. A novel functional variant in the stem cell growth factor promoter protects against severe malarial anemia. Infect Immun. 2010 Jan;78(1):453–460.
  • English M, Murphy S, Mwangi I, et al. Interobserver variation in respiratory signs of severe malaria. Arch Dis Child. 1995 Apr;72(4):334–336.
  • English M, Waruiru C, Amukoye E, et al. Deep breathing in children with severe malaria: indicator of metabolic acidosis and poor outcome. Am J Trop Med Hyg. 1996 Nov;55(5):521–524.
  • Redd SC, Bloland PB, Kazembe PN, et al. Usefulness of clinical case-definitions in guiding therapy for African children with malaria or pneumonia. Lancet. 1992 Nov 7;340(8828):1140–1143.
  • O’Dempsey TJ, McArdle TF, Laurence BE, et al. Overlap in the clinical features of pneumonia and malaria in African children. Trans R Soc Trop Med Hyg. 1993 Dec;87(6):662–665.
  • English M, Punt J, Mwangi I, et al. Clinical overlap between malaria and severe pneumonia in Africa children in hospital. Trans R Soc Trop Med Hyg. 1996 Dec;90(6):658–662.
  • Allen SJ, O’Donnell A, Alexander ND, et al. Severe malaria in children in Papua New Guinea. QJM Mon J Assoc Physicians. 1996 Oct;89(10):779–788.
  • Planche T, Agbenyega T, Bedu-Addo G, et al. A prospective comparison of malaria with other severe diseases in African children: prognosis and optimization of management. Clin Infect Dis. 2003 Oct 1;37(7):890–897.
  • Maitland K, Pamba A, Fegan G, et al. Perturbations in electrolyte levels in kenyan children with severe malaria complicated by acidosis. Clin Infect Dis. 2005 Jan 1;40(1):9–16.
  • Sasi P, Burns SP, Waruiru C, et al. Metabolic acidosis and other determinants of hemoglobin-oxygen dissociation in severe childhood Plasmodium falciparum malaria. Am J Trop Med Hyg. 2007 Aug;77(2):256–260.
  • Herdman MT, Sriboonvorakul N, Leopold SJ, et al. The role of previously unmeasured organic acids in the pathogenesis of severe malaria. Crit Care. 2015 Sep 7;19:317.
  • Leopold SJ, Ghose A, Allman EL, et al. Identifying the components of acidosis in patients with severe plasmodium falciparum malaria using metabolomics. J Infect Dis. 2019 May 5;219(11):1766–1776.
  • Berkley JA, Lowe BS, Mwangi I, et al. Bacteremia among children admitted to a rural hospital in Kenya. N Engl J Med. 2005 Jan 6;352(1):39–47.
  • Church J, Maitland K. Invasive bacterial co-infection in African children with Plasmodium falciparum malaria: a systematic review. BMC Med. 2014 Feb;19(12):31.
  • Berkley J, Mwarumba S, Bramham K, et al. Bacteraemia complicating severe malaria in children. Trans R Soc Trop Med Hyg. 1999 Jun;93(3):283–286.
  • Gómez-Pérez GP, van Bruggen R, Grobusch MP, et al. Plasmodium falciparum malaria and invasive bacterial co-infection in young African children: the dysfunctional spleen hypothesis. Malar J. 2014 Aug 26;13:335.
  • Were T, Davenport GC, Hittner JB, et al. Bacteremia in Kenyan children presenting with malaria. J Clin Microbiol. 2011 Feb;49(2):671–676.
  • Bronzan RN, Taylor TE, Mwenechanya J, et al. Bacteremia in Malawian children with severe malaria: prevalence, etiology, HIV coinfection, and outcome. J Infect Dis. 2007 Mar 15;195(6):895–904.
  • Otieno RO, Ouma C, Ong’echa JM, et al. Increased severe anemia in HIV-1-exposed and HIV-1-positive infants and children during acute malaria. AIDS. 2006 Jan 9;20(2):275–280.
  • Malamba S, Hladik W, Reingold A, et al. The effect of HIV on morbidity and mortality in children with severe malarial anaemia. Malar J. 2007 Oct 31;6(1):143.
  • Imani PD, Musoke P, Byarugaba J, et al. Human immunodeficiency virus infection and cerebral malaria in children in Uganda: a case-control study. BMC Pediatr. 2011 Jan 14;11(1):5.
  • Whitworth J, Morgan D, Quigley M, et al. Effect of HIV-1 and increasing immunosuppression on malaria parasitaemia and clinical episodes in adults in rural Uganda: a cohort study. Lancet. 2000 Sep 23;356(9235):1051–1056.
  • French N, Nakiyingi J, Lugada E, et al. Increasing rates of malarial fever with deteriorating immune status in HIV-1-infected Ugandan adults. AIDS. 2001 May 4;15(7):899–906.
  • Grimwade K, French N, Mbatha DD, et al. HIV infection as a cofactor for severe falciparum malaria in adults living in a region of unstable malaria transmission in South Africa. AIDS. 2004 Feb 20;18(3):547–554.
  • Patnaik P, Jere CS, Miller WC, et al. Effects of HIV-1 serostatus, HIV-1 RNA concentration, and CD4 cell count on the incidence of malaria infection in a cohort of adults in rural Malawi. J Infect Dis. 2005 Sep 15;192(6):984–991.
  • Cohen C, Karstaedt A, Frean J, et al. Increased prevalence of severe malaria in HIV-infected adults in South Africa. Clin Infect Dis. 2005 Dec 1;41(11):1631–1637.
  • Hendriksen ICE, Ferro J, Montoya P, et al. Diagnosis, clinical presentation, and in-hospital mortality of severe malaria in HIV-coinfected children and adults in Mozambique. Clin Infect Dis. 2012 Oct;55(8):1144–1153.
  • Hochman SE, Madaline TF, Wassmer SC, et al. Fatal pediatric cerebral malaria is associated with intravascular monocytes and platelets that are increased with HIV coinfection. mBio. 2015 Sep 22;6(5):e01390–01315.
  • van Eijk AM, Ayisi JG, Ter Kuile FO, et al. Malaria and human immunodeficiency virus infection as risk factors for anemia in infants in Kisumu, western Kenya. Am J Trop Med Hyg. 2002 Jul;67(1):44–53.
  • Angelo KM, Libman M, Caumes E, et al. Malaria after international travel: a GeoSentinel analysis, 2003–2016. Malar J. 2017 Jul 20;16(1):293.
  • WHO European health information at your fingertips. [Internet]. [cited 2019 Sep 28]. Available from: https://gateway.euro.who.int/en/indicators/hfa_330-2092-number-of-deaths-from-malaria/
  • Romi R, Boccolini D, D’Amato S, et al. Incidence of malaria and risk factors in Italian travelers to malaria endemic countries. Travel Med Infect Dis. 2010 May;8(3):144–154.
  • Checkley AM, Smith A, Smith V, et al. Risk factors for mortality from imported falciparum malaria in the United Kingdom over 20 years: an observational study. BMJ. 2012 Mar;27(344):e2116.
  • Malaria - NICE CKS [Internet]. National institute for health and care excellence. [cited 2019 Jul 28]. Available from: https://cks.nice.org.uk/malaria
  • Gérardin P, Rogier C, Ka AS, et al. Prognostic value of thrombocytopenia in African children with falciparum malaria. Am J Trop Med Hyg. 2002 Jun;66(6):686–691.
  • Obonyo CO, Vulule J, Akhwale WS, et al. In-hospital morbidity and mortality due to severe malarial anemia in western Kenya. Am J Trop Med Hyg. 2007 Dec;77(6 Suppl):23–28.
  • Lampah DA, Yeo TW, Malloy M, et al. Severe malarial thrombocytopenia: a risk factor for mortality in Papua, Indonesia. J Infect Dis. 2015 Feb 15;211(4):623–634.
  • Molyneux ME, Taylor TE, Wirima JJ, et al. Clinical features and prognostic indicators in paediatric cerebral malaria: a study of 131 comatose Malawian children. Q J Med. 1989 May;71(265):441–459.
  • Erdman LK, Petes C, Lu Z, et al. Chitinase 3-like 1 is induced by Plasmodium falciparum malaria and predicts outcome of cerebral malaria and severe malarial anaemia in a case-control study of African children. Malar J. 2014 Jul 21;13:279.
  • Conroy AL, Glover SJ, Hawkes M, et al. Angiopoietin-2 levels are associated with retinopathy and predict mortality in Malawian children with cerebral malaria: a retrospective case-control study. Crit Care Med. 2012 Mar;40(3):952–959.
  • Lovegrove FE, Tangpukdee N, Opoka RO, et al. Serum angiopoietin-1 and −2 levels discriminate cerebral malaria from uncomplicated malaria and predict clinical outcome in African children. PLoS One. 2009;4(3):e4912.
  • Lopansri BK, Anstey NM, Weinberg JB, et al. Low plasma arginine concentrations in children with cerebral malaria and decreased nitric oxide production. Lancet. 2003 Feb 22;361(9358):676–678.