Skip to main content

Inhaled nitric oxide as adjunctive therapy for severe malaria: a randomized controlled trial

Abstract

Background

Severe malaria remains a major cause of childhood mortality globally. Decreased endothelial nitric oxide is associated with severe and fatal malaria. The hypothesis was that adjunctive inhaled nitric oxide (iNO) would improve outcomes in African children with severe malaria.

Methods

A randomized, blinded, placebo-controlled trial of iNO at 80 ppm by non-rebreather mask versus room air placebo as adjunctive treatment to artesunate in children with severe malaria was conducted. The primary outcome was the longitudinal course of angiopoietin-2 (Ang-2), an endothelial biomarker of malaria severity and clinical outcome.

Results

One hundred and eighty children were enrolled; 88 were assigned to iNO and 92 to placebo (all received IV artesunate). Ang-2 levels measured over the first 72 h of hospitalization were not significantly different between groups. The mortality at 48 h was similar between groups [6/87 (6.9 %) in the iNO group vs 8/92 (8.7 %) in the placebo group; OR 0.78, 95 % CI 0.26–2.3; p = 0.65]. Clinical recovery times and parasite clearance kinetics were similar (p > 0.05). Methaemoglobinaemia >7 % occurred in 25 % of patients receiving iNO and resolved without sequelae. The incidence of neurologic deficits (<14 days), acute kidney injury, hypoglycaemia, anaemia, and haemoglobinuria was similar between groups (p > 0.05).

Conclusions

iNO at 80 ppm administered by non-rebreather mask was safe but did not affect circulating levels of Ang-2. Alternative methods of enhancing endothelial NO bioavailability may be necessary to achieve a biological effect and improve clinical outcome.

Trial Registration: ClinicalTrials.gov NCT01255215

Background

Severe malaria due to Plasmodium falciparum claims 0.6–1.2 million lives annually, 86 % of whom are children in sub-Saharan Africa [1, 2]. Despite the use of highly effective anti-malarial medications, 10–30 % patients with severe malaria will die [3], highlighting the need for new adjunctive therapy. Nitric oxide (NO), with its modulating effects on host endothelial activation, is a promising agent based on pre-clinical findings and an established safety profile in clinical practice [4, 5].

NO is a gaseous, lipid-soluble, free radical, endogenously produced from l-arginine and molecular oxygen by members of the nitric oxide synthase family [6]. NO regulates a broad range of physiologic and pathologic processes, including vasodilation, platelet aggregation, apoptosis, inflammation, chemotaxis, neurotransmission, antimicrobial defence, and endothelial activation [6].

The vascular endothelium plays a central role in the pathogenesis of severe malaria. Parasitized erythrocytes (PEs) adhere to the endothelial cells via constitutive and cytokine-inducible receptors. NO decreases endothelial cell adhesion molecule expression, and has been shown to reduce the adherence of PEs to endothelial cells [7]. Upon activation, endothelial cells release angiopoietin-2 (Ang-2) from intracellular Weibel-Palade bodies (WPB) storage granules [8]. Ang-2 functions as an autocrine regulator by sensitizing the endothelium to the effects of tumour necrosis factor, resulting in increased adhesion receptor expression [9]. Ang-2 is associated with malarial retinopathy and is an independent and quantitative marker of disease severity and clinical outcome in malaria [1012]. Ang-2 has also been used to follow disease progression and recovery in previous studies of malaria [13]. NO inhibits the exocytosis of WPB contents through S-nitrosylation of critical regulatory proteins [14].

Reduced bioavailability of NO contributes to the pathogenesis of severe malaria. African children with severe malaria have impaired production of NO [15] and low plasma arginine levels [16], the substrate for NO synthesis. Treatment of Indonesian adults with severe malaria with intravenous l-arginine increased levels of exhaled NO, and reversed malaria-associated endothelial dysfunction [17]. Pathways of endothelial activation and dysfunction are reflected in experimental models of severe malaria, where inhaled NO enhances endothelial integrity, reduces parasite accumulation in the brain vasculature, and improves survival [5, 18].

Administration of exogenous NO at 5–80 ppm is approved for use by the US FDA for neonates with hypoxic respiratory failure [19].Inhaled nitric oxide (iNO) has been safely used in clinical practice and in large clinical trials involving critically ill neonates [19], as well as in children and adults with refractory hypoxia [20]. Adverse effects attributable to iNO in these studies are rare and include methaemoglobinaemia, renal insufficiency [21] and hypotension.

To test the hypothesis that iNO would improve outcomes in children with severe malaria, a randomized, blinded, controlled trial was conducted. The primary objective was to determine if iNO at 80 ppm, relative to placebo room air, would improve endothelial function as determined by an accelerated rate of decline of Ang-2 in peripheral blood among African children with severe malaria receiving artesunate.

Methods

Trial design

This was a prospective, parallel arm, randomized, placebo-controlled, blinded trial of iNO versus placebo (1:1 ratio), among children with severe malaria, all of whom were treated with artesunate. The trial protocol has been described in detail previously [22].

Ethics, consent and permissions

The study was reviewed and approved by the Makerere University School of Medicine Research Ethics Committee (REC Protocol # 2010-107), the Uganda National Council on Science and Technology (Ref: HS 857), the National Drug Authority of Uganda (Ref: 297/ESR/NDA/DID-01/2011), and the University Health Network Research Ethics Committee, Toronto, Canada (UHN REB Number 10-0607-B). A data and safety monitoring board (DSMB) was convened and met periodically to review trial quality and adverse events. An interim analysis at the trial midpoint was conducted to review trial quality and safety, at which time the DSMB recommended that the trial proceed without modifications. The trial is registered (ClinicalTrials.gov Identifier: NCT01255215).

Setting and participants

The trial was conducted at a single centre, the Jinja Regional Referral Hospital, in Uganda. Malaria transmission is moderate and seasonal in Jinja and the surrounding Busoga catchment area [23]. The hospital operates under severe resource constraints, and over 30 % of all admissions are due to malaria.

Children (age 1–10 years) were included if they had a positive rapid diagnostic test for both P. falciparum histidine rich protein 2 (HRP2) and lactate dehydrogenase (pLDH)(First Response Malaria Ag. (pLDH/HRP2) Combo Rapid Diagnostic Test, Premier Medical Corporation Limited, India) [24], as well as selected criteria for severe malaria: repeated seizures (two or more generalized seizures in 24 h), prostration, impaired consciousness (Blantyre Coma Score <5), respiratory distress (age-related tachypnea with sustained nasal flaring, deep breathing or sub-costal retractions). Patients were not included if they had methaemoglobin (metHb) >2 % at baseline, known chronic illness (renal, cardiac or hepatic disease, diabetes, epilepsy, cerebral palsy, or AIDS), severe malnutrition (weight-for length or height below −3 standard deviations based on WHO reference charts, or symmetrical oedema involving at least the feet). Modifications to the exclusion criteria were made with regulatory committee approval after experience with the first 20 enrolled participants. The following exclusion criteria were added: haemoglobinopathy, clinical suspicion of acute bacterial meningitis, unlikely to tolerate mask for study gas delivery, and prior quinine in the emergency department. Trial nurses or clinicians from the emergency department screened patients for eligibility using a uniform checklist and clinicians made final decisions about inclusion in the study.

Randomization and blinding

In order to blind clinicians, nurses, parents, and participants to treatment while titrating and monitoring concentrations of iNO and dose-related levels of metHb and NO2, a dedicated unblinded team was used, the members of which were not involved in clinical care decisions or outcome assessments.

Eligible patients were randomly assigned to treatment with either iNO or room air placebo (both arms received intravenous artesunate). Simple randomization was employed, using a computer-generated list created by unblinded team leader (AC) prior to trial commencement. Treatment assignment was recorded on paper and kept in sequentially numbered, sealed, opaque envelopes in a locked cabinet accessible only to the unblinded study team. After patient stabilization and informed consent, the next envelope was drawn by an unblinded investigator.

iNO was indistinguishable from room air in colour and delivery apparatus (mask, tubing, a stream of vehicle air). An unblinded team member initiated the study gas while treating nurses and clinicians were out of the room. Flowmeters and monitoring devices were in locked opaque boxes accessible only to the unblinded study team. MetHb measurements were performed using non-invasive pulse CO-oximetry (Masimo Rad-57™, Masimo Corporation, Irvine, CA, USA) by unblinded study team members. All laboratory assays and statistical analyses were performed blinded to treatment allocation.

Procedures

iNO was delivered continuously at a target concentration of 80 ppm by non-rebreather mask for up to 72 h. An air compressor was used to deliver continuous flow of vehicle air, and NO from compressed cylinders was titrated into the air stream to a concentration of 80 ppm, measured continuously at the bedside using a NO-NO2 analyser (Pulmonox Sensor; Pulmonox Research and Development Corporation, Tofield, Alberta, Canada). Methaemoglobinaemia and inspired NO2 were monitored at least every 4 h. The concentration of iNO administered was adjusted downward if the metHb level in peripheral blood rose above 7 %, and was temporarily discontinued for metHb >10 %. Participants in the control group received room air by non-rebreather mask. Both groups received intravenous artesunate, the recommended first-line treatment for severe malaria, at recommended dose and frequency [25]. Follow-on oral therapy was with artemether-lumefantrine tablets or suspension for 3 days.

Bloodwork for clinical and study purposes was drawn at admission and daily during the first 72 h of hospital admission. Admission venous blood samples were analysed at the bedside for haematocrit, creatinine, lactate, and glucose [26] and at a central laboratory for parasite density, as previously described [24]. Lumbar puncture was performed at the clinician’s discretion and was analysed for cell count and differential, total protein, Gram stain and bacterial culture.

Study outcomes

The analysis was undertaken according to a pre-specified analytical plan [22]. There were no changes to any trial outcomes after the trial commenced. The primary endpoint was the longitudinal serum Ang-2 concentration over the first 72 h of hospital admission. Ang-2 was measured from serum samples using commercially available enzyme-linked immunosorbent assay (ELISA) kits (DuoSets, R&D Systems, Minneapolis, MN, USA).

Secondary trial outcomes included: mortality, recovery times, parasite clearance kinetics, and safety. Adverse events were monitored daily using paediatric toxicity tables modified from the US National Institute of Allergy and Infectious Diseases [27].

Statistical analysis

Inclusion of 180 children with severe malaria was needed to show, with 80 % power and 95 % confidence, a 50 % difference in the rate of change of Ang-2. This calculation was supported by a simulation study under various assumptions of variance and treatment effect [22].

The primary outcome, longitudinal course of Ang-2, was compared between study arms using linear mixed-effects (LME) models. All available data was used for the primary analysis. Because of (non-random) missing longitudinal data due to death, withdrawal and lost samples, sensitivity analyses were performed with different methods of adjusting for missing data (‘intention-to-treat’ analysis), as outlined in Additional file 1. Model fit was assessed by visual inspection of residuals.

For secondary binary outcomes, Chi squared or Fisher exact test were used, as appropriate. Time to event outcomes were compared with the log-rank test, and hazard ratios (HRs) together with 95 % CIs were estimated by a Cox proportional hazard model.

Statistical analyses were done with SPSS (version 16.0) and R (version 3.0.1).

Role of the funding source

The sponsor of the study had no role in study design, data collection, data analysis, data interpretation, or writing of the report.

Results

Figure 1 shows the trial profile. Recruitment occurred between 12 July, 2011 and 14 June, 2013, with last follow-up visit on 28 June, 2013. The trial ended when the pre-specified sample size was reached.

Fig. 1
figure 1

Trial profile. RDT rapid diagnostic test, Ang-2 angiopoietin-2, LME linear mixed effects

No significant differences in baseline characteristics between the two treatment groups were observed (Table 1). Ten patients were positive for both HRP2 and pLDH bands on screening rapid diagnostic test but negative on microscopy of the admission sample. One additional patient had a microscopist diagnosis of Plasmodium ovale. All these samples tested positive for P. falciparum by PCR. These cases were equally distributed between groups (5/88 (5.7 %) iNO and 6/92 (6.5 %) placebo, p = 1.0). No alternative diagnosis was apparent (blood culture negative in all cases) and two patients with negative microscopy died.

Table 1 Baseline characteristics of the two treatment groups

The median (IQR) linear rate of change of Ang-2 over the first 72 h of hospitalization was −2.1 (−3.1 to −1.2) ng/mL/day in patients receiving iNO vs −1.9 (−3.6 to −0.57) ng/mL/day in patients receiving placebo (p = 0.68). A LME model did not show a statistically significant effect of iNO on the rate of change of Ang-2 over time (p = 0.72). Figure 2a, b shows the Ang-2 levels over time for patients in the iNO and placebo groups, together with best fit curve from the LME model.

Fig. 2
figure 2

Primary biochemical trial endpoint and mortality. Longitudinal Ang-2 concentrations in treatment (iNO) and control (room air) groups did not differ significantly (p = 0.72, a and b). Solid black dots (indicating survivors), red circles (non-survivors), and best-fit curve from a LME model are shown. Survival curves were not significantly different between groups (p = 0.67, c)

Mortality outcomes (Table 2) and survival curves (Fig. 2c) show that most deaths occurred within 48 h of admission and occurred at similar frequency in both groups. Incidence of complications during hospitalization (development of coma, deterioration of coma score, new or persistent seizures, hypoglycaemia, development of severe anaemia, and haemoglobinuria) was also similar between groups (Table 2). Ang-2 levels were higher at admission among patients who subsequently died compared to survivors (median (IQR) 20 (8.8–29) ng/mL among fatal cases vs 8.9 (4.9–14) ng/mL among survivors; p = 0.0068). Co-treatments, administered by physicians blinded to randomization arm, were similar between groups, with the exception of anticonvulsants (Table 2). In particular, the use of antibiotics was not statistically significantly different between groups (82/87 (94 %) iNO and 80/91 (88 %) placebo, p = 0.19).

Table 2 Mortality, complications and co-treatments according to treatment group

Among survivors, the recovery times (time to eat, sit unsupported, localize pain, fever resolution, recovery of consciousness, and discharge) were similar between groups (Table 3). Parasite clearance kinetics with artesunate were unaffected by iNO (Table 3). No parasite recrudescence or re-infection was detected at day 14 follow-up in either group. At the time of discharge, 5/88 (5.7 %) iNO recipients and 8/92 (8.7 %) placebo recipients had neurologic deficits, including inability to sit, spastic or flaccid paresis of one or more limbs, seizures, unilateral weakness, vision loss, gaze palsy, and poor head control.

Table 3 Recovery times in surviving patients according to treatment group

Adverse events were systematically recorded using standardized tables on a daily basis in all patients (Table 4). The study gas was temporarily or permanently discontinued in 19/88 (22 %) iNO patients vs 12/92 (13 %) placebo (p = 0.13) for reasons shown in Table 5. In 5/88 (5.7 %) patients receiving iNO, study gas was temporarily discontinued because of elevated metHb levels. In all cases, iNO was resumed after metHb levels declined and iNO therapy did not need to be permanently discontinued in any patient due to recurrent or refractory methaemoglobinaemia. The study gas did not need to be titrated downward or discontinued in any patient for elevated inhalational levels of NO2.

Table 4 Adverse events according to treatment group
Table 5 Study gas discontinuation according to treatment group

Evaluation of the quality of blinding and indices of the quality of clinical care provided in the trial are provided in Additional file 1.

Discussion

iNO at 80 ppm (iNO80) by non-rebreather mask was safe but did not accelerate the decline in circulating Ang-2 levels during the first 72 h of hospitalization in this study of African children with severe malaria. No differences in clinical outcomes (e.g., mortality, recovery times) were observed, likely because a measurable biological effect on the endothelium was not achieved with this dose and route of administration of NO. Of note, parasite clearance under artesunate treatment was not affected by the addition of iNO.

Among survivors of severe malaria in this study, the rate of change of Ang-2 over the first 72 h of hospitalization was −2.2 ng/mL/day (iNO group) and −1.9 ng/mL/day (placebo group), very similar to that observed in a study involving Indonesian adults, at −2.7 ng/mL/day [13]. Among four patients who died but had repeated measurements of Ang-2, the level increased on average by +15 ng/mL/day, compared to +9.5 ng/mL/day in the study from Indonesia [13]. Moreover, elevated Ang-2 levels at admission were strongly predictive of mortality in this study and in several previous reports from several populations [12, 26, 28, 29]. These findings lend further validity to Ang-2 as a clinically informative biomarker for prognosis in children with severe malaria that can be used to follow the course of disease progression and as a surrogate endpoint for severe malaria studies.

In-hospital mortality was similar in the present trial to the largest published clinical trial of paediatric severe malaria to date, AQUAMAT [25]. In the placebo arm of the present trial (patients receiving artesunate alone) the mortality was 8.7 %, compared to 8.5 % in the artesunate arm of AQUAMAT. Mortality was similar despite evidence that patients in the present trial had more severe disease at presentation: 57 vs 32 % coma; 82 vs 30 % convulsions; 55 vs 30 % severe anaemia; 50 vs 16 % respiratory distress; 93 vs 62 % severe prostration, in the placebo arm of the present trial vs artesunate arm of AQUAMAT, respectively [25]. The clinical care in the present trial (see Additional file 1) was superior to that provided at Uganda’s national referral hospital by several indices: (1) 77 vs 23 % were seen by a clinician within one hour of presentation; (2) 68 vs 12 % received the first dose of anti-malarial drug within 2 h of presentation; (3) no missing supplies and two instances of lack of blood in the hospital compared to 50 % lacking an essential drug or supply needed for resuscitation; (4) 92 vs 29 % highest parental satisfaction rating with medical care [30].

Safety was objectively and blindly assessed using standardized toxicity tables. The only clinical sign that differed between patients receiving iNO80 and placebo was peri-orbital oedema, which resolved without discontinuation of study gas. Increased interstitial fluid may be most evident in the loose areolar tissue around the eye in a recumbent patient, and may indicate transient alterations in vascular permeability [although there was no difference in Ang-2 levels), renal dysfunction with fluid retention (although was no difference in creatinine and rates of acute kidney injury (AKI)], or other reversible disturbances of capillary hydrostatic or oncotic pressures. As expected, levels of metHb rose in patients receiving iNO80. These levels could be controlled with downward titration of the study gas and required temporary discontinuation of iNO in only 5.7 % of patients. This dose-dependent adverse event provided evidence of biological activity of the administered therapy to alter the redox state of circulating haemoglobin, although an effect on the endothelium was not observed, based on Ang-2 measurements. AKI was identified as a toxicity of iNO in a meta-analysis pooling data from multiple randomized trials involving mostly adult patients, although this effect was not evident in individual studies [21]. The rate of AKI among patients in the present study was 8.0 % in the iNO compared to 3.3 % in the placebo group, which did not represent a statistically significant difference, although the present trial, like former randomized controlled trials of iNO, may have been underpowered to detect small differences in AKI rates. Co-treatment with diazepam for acute seizure management was statistically more frequent among patients receiving iNO. The significance of this finding is unclear since the proportion of patients with new onset or persistent seizures, frequency of neurologic sequelae, and time to recovery of consciousness was similar to placebo recipients.

Efficacy of iNO80 in a murine model of experimental cerebral malaria did not translate to efficacy in humans in this clinical trial; however, pre-clinical data from this model were critical for generating the hypothesis and designing the trial. In mice, unlike humans, iNO altered Ang-2 levels, decreased blood–brain barrier dysfunction, decreased brain accumulation of parasites, and improved survival [5]. Inter-species differences in iNO80 pharmacokinetics may account for lack of efficacy in the present trial. Rapid conversion of iNO to nitrate and other stable adducts may have reduced bioavailable NO at the endothelium. On the other hand, iNO has been shown to exert pharmacological effects beyond the pulmonary vasculature in other studies in humans [31]. Although no evidence of efficacy of iNO80 was observed, other doses, routes of administration or NO donor molecules remain viable options for future investigation. Likewise, the Ang-Tie2 pathway remains a valid target for experimental interventions, and the pre-clinical murine model remains a useful tool to test novel therapy in vivo prior to clinical trials.

Conclusions

Attempts to find host-directed treatments for severe malaria over the past several decades have not yet yielded an effective adjunctive therapy. iNO80 did not affect mortality or alter the rate of change of Ang-2 in this study; however, targeting the endothelium to improve outcomes in severe malaria remains a viable strategy with broader implications for other life-threatening infections such as sepsis characterized by endothelial dysfunction. Alternative methods to increase NO bioavailability at the endothelial barrier (higher dose, donor molecules, route of delivery) deserve further investigation. Given the global burden of childhood malaria and the relatively high rate of morbidity and mortality despite treatment with anti-malarials, continued investigation of adjunctive therapy is warranted.

Abbreviations

AKI:

acute kidney injury

Ang-2:

angiopoietin-2

DSMB:

data and safety monitoring board

ELISA:

enzyme-linked immunosorbent assay

HRP2:

histidine rich protein 2

HR:

hazard ratio

iNO:

inhaled nitric oxide

IQR:

inter-quartile range

LME:

linear mixed-effects

metHb:

methaemoglobin

NO:

nitric oxide

PE:

parasitized erythrocyte

pLDH:

parasite lactate dehydrogenase

WHO:

World Health Organization

WPB:

Weibel-Palade body

References

  1. Murray CJ, Rosenfeld LC, Lim SS, Andrews KG, Foreman KJ, Haring D, et al. Global malaria mortality between 1980 and 2010: a systematic analysis. Lancet. 2012;379:413–31.

    Article  PubMed  Google Scholar 

  2. WHO. Malaria fact sheet. Geneva: World Health Organization. Available at: http://www.who.int/malaria/areas/high_risk_groups/children/en/. Accessed 7 Apr 2014.

  3. Kyu HH, Fernandez E. Artemisinin derivatives versus quinine for cerebral malaria in African children: a systematic review. Bull World Health Organ. 2009;87:896–904.

    Article  PubMed Central  PubMed  Google Scholar 

  4. Hawkes M, Opoka RO, Namasopo S, Miller C, Conroy AL, Serghides L, et al. Nitric oxide for the adjunctive treatment of severe malaria: hypothesis and rationale. Med Hypotheses. 2011;77:437–44.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  5. Serghides L, Kim H, Lu Z, Kain DC, Miller C, Francis RC, et al. Inhaled nitric oxide reduces endothelial activation and parasite accumulation in the brain, and enhances survival in experimental cerebral malaria. PLoS One. 2011;6:e27714.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Griffiths MJ, Evans TW. Inhaled nitric oxide therapy in adults. N Engl J Med. 2005;353:2683–95.

    Article  CAS  PubMed  Google Scholar 

  7. Serirom S, Raharjo WH, Chotivanich K, Loareesuwan S, Kubes P, Ho M. Anti-adhesive effect of nitric oxide on Plasmodium falciparum cytoadherence under flow. Am J Pathol. 2003;162:1651–60.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  8. Fiedler U, Scharpfenecker M, Koidl S, Hegen A, Grunow V, Schmidt JM, et al. The Tie-2 ligand angiopoietin-2 is stored in and rapidly released upon stimulation from endothelial cell Weibel-Palade bodies. Blood. 2004;103:4150–6.

    Article  CAS  PubMed  Google Scholar 

  9. Fiedler U, Reiss Y, Scharpfenecker M, Grunow V, Koidl S, Thurston G, et al. Angiopoietin-2 sensitizes endothelial cells to TNF-alpha and has a crucial role in the induction of inflammation. Nat Med. 2006;12:235–9.

    Article  CAS  PubMed  Google Scholar 

  10. Conroy AL, Glover SJ, Hawkes M, Erdman LK, Seydel KB, Taylor TE, 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;40:952–9.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  11. Erdman LK, Dhabangi A, Musoke C, Conroy AL, Hawkes M, Higgins S, et al. Combinations of host biomarkers predict mortality among Ugandan children with severe malaria: a retrospective case-control study. PLoS One. 2011;6:e17440.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  12. Lovegrove FE, Tangpukdee N, Opoka RO, Lafferty EI, Rajwans N, Hawkes M, 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:e4912.

    Article  PubMed Central  PubMed  Google Scholar 

  13. Yeo TW, Lampah DA, Gitawati R, Tjitra E, Kenangalem E, Piera K, et al. Angiopoietin-2 is associated with decreased endothelial nitric oxide and poor clinical outcome in severe falciparum malaria. Proc Natl Acad Sci USA. 2008;105:17097–102.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  14. Matsushita K, Morrell CN, Cambien B, Yang SX, Yamakuchi M, Bao C, et al. Nitric oxide regulates exocytosis by S-nitrosylation of N-ethylmaleimide-sensitive factor. Cell. 2003;115:139–50.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Anstey NM, Weinberg JB, Hassanali MY, Mwaikambo ED, Manyenga D, Misukonis MA, et al. Nitric oxide in Tanzanian children with malaria: inverse relationship between malaria severity and nitric oxide production/nitric oxide synthase type 2 expression. J Exp Med. 1996;184:557–67.

    Article  CAS  PubMed  Google Scholar 

  16. Lopansri BK, Anstey NM, Weinberg JB, Stoddard GJ, Hobbs MR, Levesque MC, et al. Low plasma arginine concentrations in children with cerebral malaria and decreased nitric oxide production. Lancet. 2003;361:676–8.

    Article  CAS  PubMed  Google Scholar 

  17. Yeo TW, Lampah DA, Gitawati R, Tjitra E, Kenangalem E, McNeil YR, et al. Impaired nitric oxide bioavailability and l-arginine reversible endothelial dysfunction in adults with falciparum malaria. J Exp Med. 2007;204:2693–704.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  18. Gramaglia I, Sobolewski P, Meays D, Contreras R, Nolan JP, Frangos JA, et al. Low nitric oxide bioavailability contributes to the genesis of experimental cerebral malaria. Nat Med. 2006;12:1417–22.

    Article  CAS  PubMed  Google Scholar 

  19. Finer NN, Barrington KJ. Nitric oxide for respiratory failure in infants born at or near term. Cochrane Database Syst Rev 2006;CD000399.

  20. Sokol J, Jacobs SE, Bohn D. Inhaled nitric oxide for acute hypoxemic respiratory failure in children and adults. Cochrane Database Syst Rev 2003; CD002787.

  21. Adhikari NK, Burns KE, Friedrich JO, Granton JT, Cook DJ, Meade MO. Effect of nitric oxide on oxygenation and mortality in acute lung injury: systematic review and meta-analysis. BMJ. 2007;334:779.

    Article  PubMed Central  PubMed  Google Scholar 

  22. Hawkes M, Opoka RO, Namasopo S, Miller C, Thorpe KE, Lavery JV, et al. Inhaled nitric oxide for the adjunctive therapy of severe malaria: protocol for a randomized controlled trial. Trials. 2011;12:176.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  23. Idro R, Aloyo J, Mayende L, Bitarakwate E, John CC, Kivumbi GW. Severe malaria in children in areas with low, moderate and high transmission intensity in Uganda. Trop Med Int Health. 2006;11:115–24.

    Article  CAS  PubMed  Google Scholar 

  24. Hawkes M, Conroy AL, Opoka RO, Namasopo S, Liles WC, John CC, et al. Use of a three-band HRP2/pLDH combination rapid diagnostic test increases diagnostic specificity for falciparum malaria in Ugandan children. Malar J. 2014;13:43.

    Article  PubMed Central  PubMed  Google Scholar 

  25. Dondorp AM, Fanello CI, Hendriksen IC, Gomes E, Seni A, Chhaganlal KD, et al. Artesunate versus quinine in the treatment of severe falciparum malaria in African children (AQUAMAT): an open-label, randomised trial. Lancet. 2010;376:1647–57.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. Hawkes M, Conroy AL, Opoka RO, Namasopo S, Liles WC, John CC, et al. Performance of point-of-care diagnostics for glucose, lactate, and hemoglobin in the management of severe malaria in a resource-constrained hospital in Uganda. Am J Trop Med Hyg. 2014;90:605–8.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  27. Little RJ, D’Agostino R, Cohen ML, Dickersin K, Emerson SS, Farrar JT, et al. The prevention and treatment of missing data in clinical trials. N Engl J Med. 2012;367:1355–60.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  28. Conroy AL, Lafferty EI, Lovegrove FE, Krudsood S, Tangpukdee N, Liles WC, et al. Whole blood angiopoietin-1 and -2 levels discriminate cerebral and severe (non-cerebral) malaria from uncomplicated malaria. Malar J. 2009;8:295.

    Article  PubMed Central  PubMed  Google Scholar 

  29. Erdman LK, Dhabangi A, Musoke C, Conroy AL, Hawkes M, Higgins S, et al. Combinations of host biomarkers predict mortality among Ugandan children with severe malaria: a retrospective case-control study. PLoS One. 2011;6:e17440.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  30. Idro R, Aloyo J. Manifestations, quality of emergency care and outcome of severe malaria in Mulago Hospital, Uganda. Afr Health Sci. 2004;4:50–7.

    PubMed Central  PubMed  Google Scholar 

  31. Wraight WM, Young JD. Renal effects of inhaled nitric oxide in humans. Br J Anaesth. 2001;86:267–9.

    Article  CAS  PubMed  Google Scholar 

  32. Mehta RL, Kellum JA, Shah SV, Molitoris BA, Ronco C, Warnock DG, et al. Acute kidney injury network: report of an initiative to improve outcomes in acute kidney injury. Crit Care. 2007;11:R31.

    Article  PubMed Central  PubMed  Google Scholar 

Download references

Authors’ contributions

MH designed the trial, directly supervised trial conduct in Uganda, managed trial patients and supervised trial participant medical care, collected the data, analysed the data, and wrote the manuscript. AC participated in the trial design, led the unblinded team (randomization, metHb monitoring), supervised the administration of iNO, performed the assays for Ang-2, and critically reviewed the manuscript. ROO participated in study design, oversaw the trial and critically reviewed the manuscript. LH participated in the trial design, supervised the administration of iNO and critically reviewed the manuscript. CM supervised the administration of iNO and performed the assays for Ang-2. HK and SH performed the assays for Ang-2. SN participated in the trial design, supervised trial participant medical care and critically reviewed the manuscript. CJ participated in the trial design, and critically reviewed the manuscript. WCL participated in the trial design and critically reviewed the manuscript. KCK conceived the study, participated in the trial design and critically reviewed the manuscript. All authors read and approved the final manuscript.

Acknowledgements

We thank all the patients and their families, the medical superintendant of the Jinja Regional Referral Hospital, the many medical officers, nurses and research assistants that cared for the patients and collected study data; the Uganda National Council on Science and Technology, the Uganda National Drug Authority, and the Data and Safety Monitoring Board for trial oversight. This study was supported by the Sandra Rotman Centre for Global Health, the Canadian Institutes of Health Research (CIHR) MOP-244701, 13721 and 136813 (KCK), Canada Research Chair in Molecular Parasitology (KCK), Canada Research Chair in Infectious Diseases and Inflammation (WCL), CIHR Clinician-Scientist Training Award (MH), and a Postdoctoral Research Award (ALC). This work was also supported by kind donations from Kim Kertland, the Tesari Foundation, and Rotary International (K-W group). The funders had no role in study design, data collection, data analysis, data interpretation, writing of the report, or decision to submit the article for publication. The researchers are independent from the funders.

Competing interests

The authors declare they have no competing interests.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kevin C. Kain.

Additional file

12936_2015_946_MOESM1_ESM.docx

Additional file 1. Sensitivity analyses and quality measures for clinical trial “Inhaled nitric oxide as adjunctive therapy for severe malaria”. Description: Sensitivity analyses on the primary and secondary (mortality) endpoints are provided. Measures of the quality of blinding and quality of clinical care in the trial are described.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hawkes, M.T., Conroy, A.L., Opoka, R.O. et al. Inhaled nitric oxide as adjunctive therapy for severe malaria: a randomized controlled trial. Malar J 14, 421 (2015). https://doi.org/10.1186/s12936-015-0946-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12936-015-0946-2

Keywords