U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

WHO Guidelines for malaria [Internet]. Geneva: World Health Organization; 2022 Feb 18.

  • This publication is provided for historical reference only and the information may be out of date.

This publication is provided for historical reference only and the information may be out of date.

Cover of WHO Guidelines for malaria

WHO Guidelines for malaria [Internet].

Show details

5CASE MANAGEMENT

Background

Malaria case management, consisting of early diagnosis and prompt effective treatment, remains a vital component of malaria control and elimination strategies. The WHO Guidelines for the treatment of malaria were first developed in 2006 and have been revised periodically, with the most recent edition published in 2015. WHO guidelines contain recommendations on clinical practice or public health policy intended to guide end-users as to the individual or collective actions that can or should be taken in specific situations to achieve the best possible health outcomes. Such recommendations are also designed to help the user to select and prioritize interventions from a range of potential alternatives. The third edition of the WHO Guidelines for the treatment of malaria consolidated here contains updated recommendations based on new evidence particularly related to dosing in children, and also includes recommendations on the use of drugs to prevent malaria in groups at high risk.

Since publication of the first edition of the Guidelines for the treatment of malaria in 2006 and the second edition in 2010, all countries in which P. falciparum malaria is endemic have progressively updated their treatment policy from use of monotherapy with drugs such as chloroquine, amodiaquine and sulfadoxine–pyrimethamine (SP) to the currently recommended artemisinin-based combination therapies (ACT). The ACTs are generally highly effective and well tolerated. This has contributed substantially to reductions in global morbidity and mortality from malaria. Unfortunately, resistance to artemisinins has arisen recently in P. falciparum in South-East Asia, which threatens these gains.

Core principles

The following core principles were used by the Guidelines Development Group that drew up the Guidelines for the Treatment of Malaria.

1. Early diagnosis and prompt, effective treatment of malaria

Uncomplicated falciparum malaria can progress rapidly to severe forms of the disease, especially in people with no or low immunity, and severe falciparum malaria is almost always fatal without treatment. Therefore, programmes should ensure access to early diagnosis and prompt, effective treatment within 24–48 h of the onset of malaria symptoms.

2. Rational use of antimalarial agents

To reduce the spread of drug resistance, limit unnecessary use of antimalarial drugs and better identify other febrile illnesses in the context of changing malaria epidemiology, antimalarial medicines should be administered only to patients who truly have malaria. Adherence to a full treatment course must be promoted. Universal access to parasitological diagnosis of malaria is now possible with the use of quality-assured rapid diagnostic tests (RDTs), which are also appropriate for use in primary health care and community settings.

3. Combination therapy

Preventing or delaying resistance is essential for the success of both national and global strategies for control and eventual elimination of malaria. To help protect current and future antimalarial medicines, all episodes of malaria should be treated with at least two effective antimalarial medicines with different mechanisms of action (combination therapy).

4. Appropriate weight-based dosing

To prolong their useful therapeutic life and ensure that all patients have an equal chance of being cured, the quality of antimalarial drugs must be ensured, and antimalarial drugs must be given at optimal dosages. Treatment should maximize the likelihood of rapid clinical and parasitological cure and minimize transmission from the treated infection. To achieve this, dosage regimens should be based on the patient’s weight and should provide effective concentrations of antimalarial drugs for a sufficient time to eliminate the infection in all target populations.

Please refer to Malaria case management: operations manual (110).

5.1. Diagnosing malaria (2015)

Suspected malaria

The signs and symptoms of malaria are non-specific. Malaria is suspected clinically primarily on the basis of fever or a history of fever. There is no combination of signs or symptoms that reliably distinguishes malaria from other causes of fever; diagnosis based only on clinical features has very low specificity and results in overtreatment. Other possible causes of fever and whether alternative or additional treatment is required must always be carefully considered. The focus of malaria diagnosis should be to identify patients who truly have malaria, to guide rational use of antimalarial medicines.

In malaria-endemic areas, malaria should be suspected in any patient presenting with a history of fever or temperature ≥ 37.5 °C and no other obvious cause. In areas in which malaria transmission is stable (or during the high-transmission period of seasonal malaria), malaria should also be suspected in children with palmar pallor or a haemoglobin concentration of < 8 g/dL. High-transmission settings include many parts of sub-Saharan Africa and some parts of Oceania.

In settings where the incidence of malaria is very low, parasitological diagnosis of all cases of fever may result in considerable expenditure to detect only a few patients with malaria. In these settings, health workers should be trained to identify patients who may have been exposed to malaria (e.g. recent travel to a malaria-endemic area without protective measures) and have fever or a history of fever with no other obvious cause, before they conduct a parasitological test.

In all settings, suspected malaria should be confirmed with a parasitological test. The results of parasitological diagnosis should be available within a short time (< 2 h) of the patient presenting. In settings where parasitological diagnosis is not possible, a decision to provide antimalarial treatment must be based on the probability that the illness is malaria.

In children < 5 years, the practical algorithms for management of the sick child provided by the WHO–United Nations Children’s Fund (UNICEF) strategy for Integrated Management of Childhood Illness (111) should be used to ensure full assessment and appropriate case management at first-level health facilities and at the community level.

Parasitological diagnosis

The benefit of parasitological diagnosis relies entirely on an appropriate management response of health care providers. The two methods used routinely for parasitological diagnosis of malaria are light microscopy and immunochromatographic RDTs. The latter detect parasite-specific antigens or enzymes that are either genus or species specific.

Both microscopy and RDTs must be supported by a quality assurance programme. Antimalarial treatment should be limited to cases with positive tests, and patients with negative results should be reassessed for other common causes of fever and treated appropriately.

In nearly all cases of symptomatic malaria, examination of thick and thin blood films by a competent microscopist will reveal malaria parasites. Malaria RDTs should be used if quality-assured malaria microscopy is not readily available. RDTs for detecting PfHRP2 can be useful for patients who have received incomplete antimalarial treatment, in whom blood films can be negative. This is particularly likely if the patient received a recent dose of an artemisinin derivative. If the initial blood film examination is negative in patients with manifestations compatible with severe malaria, a series of blood films should be examined at 6–12 h intervals, or an RDT (preferably one detecting PfHRP2) should be performed. If both the slide examination and the RDT results are negative, malaria is extremely unlikely, and other causes of the illness should be sought and treated.

This document does not include recommendations for use of specific RDTs or for interpreting test results. For guidance, see the WHO manual Universal access to malaria diagnostic testing (112).

Diagnosis of malaria

In patients with suspected severe malaria and in other high-risk groups, such as patients living with HIV/AIDS, absence or delay of parasitological diagnosis should not delay an immediate start of antimalarial treatment.

At present, molecular diagnostic tools based on nucleic-acid amplification techniques (e.g. loop-mediated isothermal amplification or PCR) do not have a role in the clinical management of malaria.

Where P. vivax malaria is common and microscopy is not available, it is recommended that a combination RDT be used that allows detection of P. vivax (pLDH antigen from P. vivax) or pan-malarial antigens (Pan-pLDH or aldolase).

Light microscopy

Microscopy not only provides a highly sensitive, specific diagnosis of malaria when performed well but also allows quantification of malaria parasites and identification of the infecting species. Light microscopy involves relatively high costs for training and supervision, and the accuracy of diagnosis is strongly dependent on the competence of the microscopist. Microscopy technicians may also contribute to the diagnosis of non-malarial diseases.

Although nucleic acid amplification-based tests are more sensitive, light microscopy is still considered the “field standard” against which the sensitivity and specificity of other methods must be assessed. A skilled microscopist can detect asexual parasites at a density of < 10 per µL of blood, but under typical field conditions, the limit of sensitivity is approximately 100 parasites per µL (113). This limit of detection approximates the lower end of the pyrogenic density range. Thus, microscopy provides good specificity for diagnosing malaria as the cause of a presenting febrile illness. More sensitive methods allow detection of an increasing proportion of cases of incidental parasitaemia in endemic areas, thus reducing the specificity of a positive test. Light microscopy has other important advantages:

  • low direct costs, if laboratory infrastructure to maintain the service is available;
  • high sensitivity, if the performance of microscopy is high;
  • differentiation of Plasmodia species;
  • determination of parasite densities – notably identification of hyperparasitaemia;
  • detection of gametocytaemia;
  • allows monitoring of responses to therapy and
  • can be used to diagnose many other conditions.

Good performance of microscopy can be difficult to maintain, because of the requirements for adequate training and supervision of laboratory staff to ensure competence in malaria diagnosis, electricity, good quality slides and stains, provision and maintenance of good microscopes and maintenance of quality assurance (114) and control of laboratory services [94][95].

Numerous attempts have been made to improve malaria microscopy, but none has proven to be superior to the classical method of Giemsa staining and oil-immersion microscopy for performance in typical health care settings (115).

Rapid diagnostic tests

Rapid diagnostic tests (RDTs) are immuno-chromatographic tests for detecting parasite-specific antigens in a finger-prick blood sample. Some tests allow detection of only one species (P. falciparum); others allow detection of one or more of the other species of human malaria parasites (P. vivax, P. malariae and P. ovale) (116) (117)(118). They are available commercially in various formats, e.g. dipsticks, cassettes and cards. Cassettes and cards are easier to use in difficult conditions outside health facilities. RDTs are relatively simple to perform and to interpret, and they do not require electricity or special equipment (119).

Since 2012, WHO has recommended that RDTs should be selected in accordance with the following criteria, based on the results of the assessments of the WHO Malaria RDT Product Testing programme (120):

  • For detection of P. falciparum in all transmission settings, the panel detection score against P. falciparum samples should be at least 75% at 200 parasites/µL.
  • For detection of P. vivax in all transmission settings the panel detection score against P. vivax samples should be at least 75% at 200 parasites/µL.
  • The false positive rate should be less than 10%.
  • The invalid rate should be less than 5%.

Current tests are based on the detection of histidine-rich protein 2 (HRP2), which is specific for P. falciparum, pan-specific or species-specific Plasmodium lactate dehydrogenase (pLDH) or pan-specific aldolase. The different characteristics of these antigens may affect their suitability for use in different situations, and these should be taken into account in programmes for RDT implementation. The tests have many potential advantages, including:

  • rapid provision of results and extension of diagnostic services to the lowest-level health facilities and communities;
  • fewer requirements for training and skilled personnel (for instance, a general health worker can be trained in 1 day); and
  • reinforcement of patient confidence in the diagnosis and in the health service in general.

They also have potential disadvantages, including:

  • inability, in the case of PfHRP2-based RDTs, to distinguish new infections from recently and effectively treated infections, due to the persistence of PfHRP2 in the blood for 1–5 weeks after effective treatment;
  • the presence in countries in the Amazon region of variable frequencies of HRP2 deletions in P. falciparum parasites, making HRP2-based tests not suitable in this region (121);
  • poor sensitivity for detecting P. malariae and P. ovale; and
  • the heterogeneous quality of commercially available products and the existence of lot-to-lot variation.

In a systematic review (122), the sensitivity and specificity of RDTs in detecting P. falciparum in blood samples from patients in endemic areas attending ambulatory health facilities with symptoms suggestive of malaria were compared with the sensitivity and specificity of microscopy or polymerase chain reaction. The average sensitivity of PfHRP2-detecting RDTs was 95.0% (95% confidence interval [CI], 93.5–96.2%), and the specificity was 95.2% (93.4–99.4%). RDTs for detecting pLDH from P. falciparum are generally less sensitive and more specific than those for detecting HRP2, with an average sensitivity (95% CI) of 93.2% (88.0–96.2%) and a specificity of 98.5% (96.7–99.4%). Several studies have shown that health workers, volunteers and private sector providers can, with adequate training and supervision, use RDTs correctly and provide accurate malaria diagnoses. The criteria for selecting RDTs or microscopy can be found in the WHO Recommended selection criteria for the procurement of malaria rapid diagnostic tests (123).

Diagnosis with either microscopy or RDTs is expected to reduce overuse of antimalarial medicines by ensuring that treatment is given only to patients with confirmed malaria infection, as opposed to treating all patients with fever (124). Although providers of care may be willing to perform diagnostic tests, they do not, however, always respond appropriately to the results. This is especially true when they are negative. It is therefore important to ensure the accuracy of parasite- based diagnosis and also to demonstrate this to users and to provide them with the resources to manage both positive and negative results adequately (112).

Immunodiagnosis and nucleic acid amplification test methods

Detection of antibodies to parasites, which may be useful for epidemiological studies, is neither sensitive nor specific enough to be of use in the management of patients suspected of having malaria (125).

Techniques to detect parasite nucleic acid, e.g. polymerase chain reaction and loop-mediated isothermal amplification, are highly sensitive and very useful for detecting mixed infections, in particular at low parasite densities that are not detectable by conventional microscopy or with RDTs. They are also useful for studies of drug resistance and other specialized epidemiological investigations (126); however, they are not generally available for large-scale field use in malaria- endemic areas, nor are they appropriate for routine diagnosis in endemic areas where a large proportion of the population may have low-density parasitaemia.

These techniques may be useful for population surveys and focus investigation in malaria elimination programmes.

At present, nucleic acid-based amplification techniques have no role in the clinical management of malaria or in routine surveillance systems (127).

Good practice statement

All cases of suspected malaria should have a parasitological test (microscopy or RDT) to confirm the diagnosis.

Both microscopy and RDTs should be supported by a quality assurance programme.

Justification

Prompt, accurate diagnosis of malaria is part of effective disease management. All patients with suspected malaria should be treated on the basis of a confirmed diagnosis by microscopy examination or RDT testing of a blood sample. Correct diagnosis in malaria-endemic areas is particularly important for the most vulnerable population groups, such as young children and non-immune populations, in whom falciparum malaria can be rapidly fatal. High specificity will reduce unnecessary treatment with antimalarial drugs and improve the diagnosis of other febrile illnesses in all settings.

WHO strongly advocates a policy of “test, treat and track” to improve the quality of care and surveillance.

5.2. Treating uncomplicated malaria

Definition of uncomplicated malaria

A patient who presents with symptoms of malaria and a positive parasitological test (microscopy or RDT) but with no features of severe malaria is defined as having uncomplicated malaria (see section 7.1 for definition of severe malaria).

Therapeutic objectives

The clinical objectives of treating uncomplicated malaria are to cure the infection as rapidly as possible and to prevent progression to severe disease. “Cure” is defined as elimination of all parasites from the body. The public health objectives of treatment are to prevent onward transmission of the infection to others and to prevent the emergence and spread of resistance to antimalarial drugs.

Incorrect approaches to treatment

Use of monotherapy

The continued use of artemisinins or any of the partner medicines alone will compromise the value of ACT by selecting for drug resistance.

As certain patient groups, such as pregnant women, may need specifically tailored combination regimens, single artemisinin derivatives will still be used in selected referral facilities in the public sector, but they should be withdrawn entirely from the private and informal sectors and from peripheral public health care facilities.

Similarly, continued availability of amodiaquine, mefloquine and SP as monotherapies in many countries is expected to shorten their useful therapeutic life as partner drugs of ACT, and they should be withdrawn wherever possible.

Incomplete dosing

In endemic regions, some semi-immune malaria patients are cured by an incomplete course of antimalarial drugs or by a treatment regimen that would be ineffective in patients with no immunity. In the past, this led to different recommendations for patients considered semi-immune and those considered non-immune. As individual immunity can vary considerably, even in areas of moderate-to-high transmission intensity, this practice is no longer recommended. A full treatment course with a highly effective ACT is required whether or not the patient is considered to be semi-immune.

Another potentially dangerous practice is to give only the first dose of a treatment course to patients with suspected but unconfirmed malaria, with the intention of giving the full treatment if the diagnosis is confirmed. This practice is unsafe, could engender resistance, and is not recommended.

Additional considerations for clinical management

Can the patient take oral medication?

Some patients cannot tolerate oral treatment and will require parenteral or rectal administration for 1–2 days, until they can swallow and retain oral medication reliably. Although such patients do not show other signs of severity, they should receive the same initial antimalarial treatments recommended for severe malaria. Initial rectal or parenteral treatment must always be followed by a full 3-day course of ACT.

Use of antipyretics

In young children, high fevers are often associated with vomiting, regurgitation of medication and seizures. They are thus treated with antipyretics and, if necessary, fanning and tepid sponging. Antipyretics should be used if the core temperature is > 38.5 °C. Paracetamol (acetaminophen) at a dose of 15 mg/kg bw every 4 h is widely used; it is safe and well tolerated and can be given orally or as a suppository. Ibuprofen (5 mg/kg bw) has been used successfully as an alternative in the treatment of malaria and other childhood fevers, but, like aspirin and other non-steroidal anti-inflammatory drugs, it is no longer recommended because of the risks of gastrointestinal bleeding, renal impairment and Reye’s syndrome.

Use of anti-emetics

Vomiting is common in acute malaria and may be severe. Parenteral antimalarial treatment may therefore be required until oral administration is tolerated. Then a full 3-day course of ACT should be given. Anti-emetics are potentially sedative and may have neuropsychiatric adverse effects, which could mask or confound the diagnosis of severe malaria. They should therefore be used with caution.

Management of seizures

Generalized seizures are more common in children with P. falciparum malaria than in those with malaria due to other species. This suggests an overlap between the cerebral pathology resulting from falciparum malaria and febrile convulsions. As seizures may be a prodrome of cerebral malaria, patients who have more than two seizures within a 24 h period should be treated as for severe malaria. If the seizures continue, the airways should be maintained and anticonvulsants given (parenteral or rectal benzodiazepines or intramuscular paraldehyde). When the seizure has stopped, the child should be treated as indicated in section 7.10.5, if his or her core temperature is > 38.5 °C. There is no evidence that prophylactic anticonvulsants are beneficial in otherwise uncomplicated malaria, and they are not recommended.

5.2.1. Artemisinin-based combination therapy

Strong recommendation for, High certainty evidence

Treat children and adults with uncomplicated P. falciparum malaria (except pregnant women in their first trimester) with one of the following ACTs:

  • artemether + lumefantrine
  • artesunate + amodiaquine
  • artesunate + mefloquine
  • dihydroartemisinin + piperaquine
  • artesunate + sulfadoxine–pyrimethamine (SP)
  • artesunate + pyronaridine (currently unGRADEd, anticipated to be updated in 2022)

Artesunate pyronaridine is included in the WHO list of prequalified medicines for malaria, the Model List of Essential Medicines and the Model List of Medicines for Children. The drug has also received a positive scientific opinion from the European Medicines Agency and undergone a positive review by the WHO Advisory Committee on Safety of Medicinal Products. Countries can consider including this medicine in their national treatment guidelines for the treatment of malaria based on WHO’s position on the use of this drug pending the formal recommendation anticipated in 2021. WHO’s position was published in the information note The use of artesunate-pyronaridine for the treatment of uncomplicated malaria (122) which clarifies that artesunate pyronaridine can be considered a safe and efficacious ACT for the treatment of uncomplicated malaria in adults and children weighing 5 kg and over in all malaria-endemic areas.

Practical Info

The pipeline for new antimalarial drugs is healthier than ever before, and several new compounds are in various stages of development. Some novel antimalarial agents are already registered in some countries. The decision to recommend antimalarial drugs for general use depends on the strength of the evidence for safety and efficacy and the context of use. In general, when there are no satisfactory alternatives, newly registered drugs may be recommended; however, for global or unrestricted recommendations, considerably more evidence than that submitted for registration is usually required, to provide sufficient confidence for their safety, efficacy and relative merits as compared with currently recommended treatments.

Several new antimalarial drugs or new combinations have been introduced recently. Some are still in the pre-registration phase and are not discussed here. Arterolane + piperaquine, artemisinin + piperaquine base and artemisinin + napththoquine are new ACTs, which are registered and used in some countries. In addition, there are several new generic formulations of existing drugs. None of these yet has a sufficient evidence base for general recommendation (i.e. unrestricted use).

Artesunate + pyronaridine

A systematic review of artesunate + pyronaridine included six trials with a total of 3718 patients. Artesunate + pyronaridine showed good efficacy as compared with artemether + lumefantrine and artesunate + mefloquine in adults and older children with P. falciparum malaria, but the current evidence for young children is insufficient to be confident that the drug is as effective as currently recommended options. In addition, regulatory authorities noted slightly higher hepatic transaminase concentrations in artesunate + pyronaridine recipients than in comparison groups and recommended further studies to characterize the risk for hepatotoxicity. Preliminary data from repeat-dosing studies are reassuring.

In 2012, artesunate-pyronaridine was granted a positive scientific opinion under the European Medicines Agency (EMA) Article 58 procedure, but with a restricted label, mainly due to concerns over potential hepatotoxicity of the pyronaridine component, efficacy in children under 5 years of age, and safety, especially with repeat dosing (126). In 2015, an EMA Scientific Advisory Group concluded that cumulative safety data on hepatic events had provided sufficient evidence to alleviate concerns over hepatotoxicity and thus to allow recommendation of the use of artesunate pyronaridine for the treatment and re-treatment of uncomplicated malaria in patients without signs of hepatic injury (including children weighing 5 kg and over).

The EMA therefore modified the product label to remove all restrictions on repeat dosing, on use only in areas of high antimalarial drug resistance and low malaria transmission, and on requirements to monitor liver function. In addition, it granted a positive scientific opinion for artesunate-pyronaridine granules for the treatment of children with a body weight of 5–20 kg (125). Artesunate-pyronaridine was included in WHO’s list of prequalified medicines for malaria in April 2012, based on the EMA’s positive scientific opinion of this product in accordance with Article 58. Since labelling provisions are based on EMA conclusions, these provisions were updated as a result of the EMA’s 2015 review. Products included in the WHO prequalification list are those that have been assessed through the various mechanisms and found to comply with WHO-recommended regulatory standards and requirements for quality, safety and efficacy.

In June 2017, artesunate-pyronaridine was also added to the WHO Model List of Essential Medicines and Model List of Essential Medicines for Children. Due to the hepatotoxicity concerns identified in 2012, the WHO Guidelines for the treatment of malaria (2015) did not recommend the use of artesunate-pyronaridine for general use. A further meeting in December 2017 resulted in the need for GMP to request, in 2018, the support of the WHO Advisory Committee on Safety of Medicinal Products to conduct an independent expert review of all available data and information. Having completed its review, the committee considered that the current safety restrictions on the use of artesunate-pyronaridine (Pyramax®) for the treatment of uncomplicated malaria, as stated in the Guidelines for the treatment of malaria, are no longer justified (126). GMP will revise the Guidelines based on new information available in 2021.

Arterolane + piperaquine is a combination of a synthetic ozonide and piperaquine phosphate that is registered in India. There are currently insufficient data to make general recommendations.

Artemisinin + piperaquine base combines two well-established, well-tolerated compounds. It differs from previous treatments in that the piperaquine is in the base form, the artemisinin dose is relatively low, and the current recommendation is for only a 2-day regimen. There are insufficient data from clinical trials for a general recommendation, and there is concern that the artemisinin dose regimen provides insufficient protection against resistance to the piperaquine component.

Artemisinin + naphthoquine is also a combination of two relatively old compounds that is currently being promoted as a single-dose regimen, contrary to WHO advice for 3 days of the artemisinin derivative. There are currently insufficient data from rigorously conducted randomized controlled trials to make general recommendations.

Many ACTs are generics. The bioavailability of generics of currently recommended drugs must be comparable to that of the established, originally registered product, and the satisfactory pharmaceutical quality of the product must be maintained.

Please refer to Good procurement practices for artemisinin-based antimalaria medicines (127).

Evidence To Decision

Benefits and harms

Recommendation: Treat adults and children with uncomplicated P. falciparum malaria (including infants, pregnant women in their second and third trimesters and breastfeeding women) with ACT.

Desirable effects

  • Studies have consistently demonstrated that the five WHO-recommended ACTs result in < 5% PCR-adjusted treatment failures in settings with no resistance to the partner drug (high- quality evidence).

Undesirable effects

  • Increased cost.

Recommendation: Dihydroartemisinin + piperaquine is recommended for general use.

Desirable effects:

  • A PCR-adjusted treatment failure rate of < 5% has been seen consistently in trials of dihydroartemisinin + piperaquine (high-quality evidence).
  • Dihydroartemisinin + piperaquine has a longer half-life than artemether + lumefantrine, and fewer new infections occur within 9 weeks of treatment with dihydroartemisinin + piperaquine (high-quality evidence).
  • Dihydroartemisinin + piperaquine and artesunate + mefloquine have similar half-lives, and a similar frequency of new infections is seen within 9 weeks of treatment (moderate-quality evidence).

Undesirable effects:

  • A few more patients receiving dihydroartemisinin + piperaquine than those given artesunate + mefloquine had a prolonged QT interval (low-quality evidence)
  • A few more patients receiving dihydroartemisinin + piperaquine than those given artesunate + mefloquine or artemether + lumefantrine had borderline QT prolongation.

Certainty of the Evidence Image ch6if1.jpg

For all critical outcomes: High.

Preference and values

Justification

GRADE

In the absence of resistance to the partner drug, the five recommended ACTs have all been shown to achieve a PCR-adjusted treatment failure rate of 5% in many trials in several settings in both adults and children (high-quality evidence) (123)(124).

Other considerations

The guideline development group decided to recommend a menu of approved combinations, from which countries can select first- and second-line treatment.

Remarks

Recommendation: Treat adults and children with uncomplicated P. falciparum malaria (including infants, pregnant women in their second and third trimesters and breastfeeding women) with ACT.

The WHO-approved first-line ACT options are: artemether + lumefantrine, artesunate + amodiaquine, artesunate + mefloquine, dihydroartemisinin + piperaquine and artesunate + sulfadoxine–pyrimethamine.

These options are recommended for adults and children, including infants, lactating women and pregnant women in their second and third trimester.

In deciding which ACTs to adopt in national treatment policies, national policy- makers should take into account: the pattern of resistance to antimalarial drugs in the country, the relative efficacy and safety of the combinations, their cost, the availability of paediatric formulations and the availability of co-formulated products.

Fixed-dose combinations are preferred to loose tablets or co-blistered products.

The Guideline Development Group decided to recommend a “menu” of approved combinations from which countries can select first- and second- line therapies. Modelling studies suggest that having multiple first-line ACTs available for use may help to prevent or delay the development of resistance.

Recommendation: Dihydroartemisinin + piperaquine is recommended for general use.

A systematic review showed that the dosing regimen of dihydroartemisinin + piperaquine currently recommended by the manufacturers leads to sub-optimal dosing in young children. The group plans to recommend a revised dosing regimen based on models of pharmacokinetics.

Further studies of the risk for QT interval prolongation have been requested by the European Medicines Agency.

ACT is a combination of a rapidly acting artemisinin derivative with a longer-acting (more slowly eliminated) partner drug. The artemisinin component rapidly clears parasites from the blood (reducing parasite numbers by a factor of approximately 10 000 in each 48 h asexual cycle) and is also active against the sexual stages of the gametocytes that mediate onward transmission to mosquitos. The longer- acting partner drug clears the remaining parasites and provides protection against development of resistance to the artemisinin derivative. Partner drugs with longer elimination half-lives also provide a period of post-treatment prophylaxis.

The GDG recommended dihydroartemisinin + piperaquine for use in 2009 but re-evaluated the evidence in 2013 because additional data on its safety had become available. The group noted the small absolute prolongation of the QT interval with dihydroartemisinin + piperaquine but was satisfied that the increase was of comparable magnitude to that observed with chloroquine and was not important clinically (127)(128).

5.2.2. Duration of treatment

A 3-day course of the artemisinin component of ACTs covers two asexual cycles, ensuring that only a small fraction of parasites remain for clearance by the partner drug, thus reducing the potential development of resistance to the partner drug. Shorter courses (1–2 days) are therefore not recommended, as they are less effective, have less effect on gametocytes and provide less protection for the slowly eliminated partner drug.

Treating uncomplicated P. falciparum malaria (2015)

Strong recommendation for, High certainty evidence

Duration of ACT treatment: ACT regimens should provide 3 days’ treatment with an artemisinin derivative.

Evidence To Decision

Benefits and harms

Desirable effects

  • Fewer patients taking ACTs containing 3 days of an artemisinin derivative experience treatment failure within the first 28 days (high-quality evidence).
  • Fewer participants taking ACTs containing 3 days of an artemisinin derivative have gametocytaemia at day 7 (high-quality evidence).
Certainty of the Evidence Image ch6if1.jpg

For all critical outcomes: High.

Preference and values

Justification

GRADE

In four randomized controlled trials in which the addition of 3 days of artesunate to SP was compared directly with 1 day of artesunate with SP:

  • Three days of artesunate reduced the PCR-adjusted treatment failure rate within the first 28 days from that with 1 day of artesunate (RR, 0.45; 95% CI, 0.36–0.55, four trials, 1202 participants, high-quality evidence).
  • Three days of artesunate reduced the number of participants who had gametocytaemia at day 7 from that with 1 day of artesunate (RR, 0.74; 95% CI, 0.58–0.93, four trials, 1260 participants, high-quality evidence).

Other considerations

The guideline development group considered that 3 days of artemisinin derivative are necessary to provide sufficient efficacy, promote good adherence and minimize the risk of drug resistance resulting from incomplete treatment.

Remarks

Longer ACT treatment may be required to achieve > 90% cure rate in areas with artemisinin-resistant P. falciparum, but there are insufficient trials to make definitive recommendations. A 3-day course of the artemisinin component of ACTs covers two asexual cycles, ensuring that only a small fraction of parasites remain for clearance by the partner drug, thus reducing the potential development of resistance to the partner drug. Shorter courses (1–2 days) are therefore not recommended, as they are less effective, have less effect on gametocytes and provide less protection for the slowly eliminated partner drug.

Rationale for the recommendation

The Guideline Development Group considers that 3 days of an artemisinin derivative are necessary to provide sufficient efficacy, promote good adherence and minimize the risk for drug resistance due to incomplete treatment.

5.2.3. Dosing of ACTS

ACT regimens must ensure optimal dosing to prolong their useful therapeutic life, i.e. to maximize the likelihood of rapid clinical and parasitological cure, minimize transmission and retard drug resistance.

It is essential to achieve effective antimalarial drug concentrations for a sufficient time (exposure) in all target populations in order to ensure high cure rates. The dosage recommendations below are derived from understanding the relationship between dose and the profiles of exposure to the drug (pharmacokinetics) and the resulting therapeutic efficacy (pharmacodynamics) and safety. Some patient groups, notably younger children, are not dosed optimally with the “dosage regimens recommended by manufacturers, which compromises efficacy and fuels resistance. In these guidelines when there was pharmacological evidence that certain patient groups are not receiving optimal doses, dose regimens were adjusted to ensure similar exposure across all patient groups.

Weight-based dosage recommendations are summarized below. While age-based dosing may be more practical in children, the relation between age and weight differs in different populations. Age-based dosing can therefore result in under- dosing or over-dosing of some patients, unless large, region-specific weight-for-age databases are available to guide dosing in that region.

Factors other than dosage regimen may also affect exposure to a drug and thus treatment efficacy. The drug exposure of an individual patient also depends on factors such as the quality of the drug, the formulation, adherence and, for some drugs, co-administration with fat. Poor adherence is a major cause of treatment failure and drives the emergence and spread of drug resistance. Fixed-dose combinations encourage adherence and are preferred to loose (individual) tablets. Prescribers should take the time necessary to explain to patients why they should complete antimalarial course.

Artemether + lumefantrine

Formulations currently available: Dispersible or standard tablets containing 20 mg artemether and 120 mg lumefantrine, and standard tablets containing 40 mg artemether and 240 mg lumefantrine in a fixed-dose combination formulation. The flavoured dispersible tablet paediatric formulation facilitates use in young children.

Target dose range: A total dose of 5–24 mg/kg bw of artemether and 29–144 mg/ kg bw of lumefantrine

Recommended dosage regimen: Artemether + lumefantrine is given twice a day for 3 days (total, six doses). The first two doses should, ideally, be given 8 h apart.

Body weight (kg)Dose (mg) of artemether + lumefantrine given twice daily for 3 days
5 to < 1520 + 120
15 to < 2540 + 240
25 to < 3560 + 360
≥ 3580 + 480

Factors associated with altered drug exposure and treatment response:

  • Decreased exposure to lumefantrine has been documented in young children (<3 years) as well as pregnant women, large adults, patients taking mefloquine, rifampicin or efavirenz and in smokers. As these target populations may be at increased risk for treatment failure, their responses to treatment should be monitored more closely and their full adherence ensured.
  • Increased exposure to lumefantrine has been observed in patients concomitantly taking lopinavir- lopinavir/ritonavir-based antiretroviral agents but with no increase in toxicity; therefore, no dosage adjustment is indicated.

Additional comments:

  • An advantage of this ACT is that lumefantrine is not available as a monotherapy and has never been used alone for the treatment of malaria.
  • Absorption of lumefantrine is enhanced by co-administration with fat. Patients or caregivers should be informed that this ACT should be taken immediately after food or a fat containing drink (e.g. milk), particularly on the second and third days of treatment.

Artesunate + amodiaquine

Formulations currently available: A fixed-dose combination in tablets containing 25 + 67.5 mg, 50 + 135 mg or 100 + 270 mg of artesunate and amodiaquine, respectively

Target dose and range: The target dose (and range) are 4 (2–10) mg/kg bw per day artesunate and 10 (7.5–15) mg/kg bw per day amodiaquine once a day for 3 days. A total therapeutic dose range of 6–30 mg/kg bw per day artesunate and 22.5–45 mg/kg bw per dose amodiaquine is recommended.

Body weight (kg)Artesunate + amodiaquine dose (mg) given daily for 3 days
4.5 to < 925 + 67.5
9 to < 1850 + 135
18 to < 36100 + 270
≥ 36200 + 540

Factors associated with altered drug exposure and treatment response:

  • Treatment failure after amodiaquine monotherapy was more frequent among children who were underweight for their age. Therefore, their response to artesunate + amodiaquine treatment should be closely monitored.
  • Artesunate + amodiaquine is associated with severe neutropenia, particularly in patients co-infected with HIV and especially in those on zidovudine and/or cotrimoxazole. Concomitant use of efavirenz increases exposure to amodiaquine and hepatotoxicity. Thus, concomitant use of artesunate + amodiaquine by patients taking zidovudine, efavirenz and cotrimoxazole should be avoided, unless this is the only ACT promptly available.

Additional comments:

  • No significant changes in the pharmacokinetics of amodiaquine or its metabolite desethylamodiaquine have been observed during the second and third trimesters of pregnancy; therefore, no dosage adjustments are recommended.
  • No effect of age has been observed on the plasma concentrations of amodiaquine and desethylamodiaquine, so no dose adjustment by age is indicated. Few data are available on the pharmacokinetics of amodiaquine in the first year of life.

Artesunate + mefloquine

Formulations currently available: A fixed-dose formulation of paediatric tablets containing 25 mg artesunate and 55 mg mefloquine hydrochloride (equivalent to 50 mg mefloquine base) and adult tablets containing 100 mg artesunate and 220 mg mefloquine hydrochloride (equivalent to 200 mg mefloquine base)

Target dose and range: Target doses (ranges) of 4 (2–10) mg/kg bw per day artesunate and 8.3 (7–11) mg/kg bw per day mefloquine, given once a day for 3 days

Body weight (kg)Artesunate + mefloquine dose (mg) given daily for 3 days
5 to < 925 + 55
9 to < 1850 + 110
18 to < 30100 + 220
≥ 30200 + 440

Additional comments:

  • Mefloquine was associated with increased incidences of nausea, vomiting, dizziness, dysphoria and sleep disturbance in clinical trials, but these symptoms are seldom debilitating, and, where this ACT has been used, it has generally been well tolerated. To reduce acute vomiting and optimize absorption, the total mefloquine dose should preferably be split over 3 days, as in current fixed-dose combinations.
  • As concomitant use of rifampicin decreases exposure to mefloquine, potentially decreasing its efficacy, patients taking this drug should be followed up carefully to identify treatment failures.

Artesunate + sulfadoxine–pyrimethamine

Formulations: Currently available as blister-packed, scored tablets containing 50 mg artesunate and fixed dose combination tablets comprising 500 mg sulfadoxine + 25 mg pyrimethamine. There is no fixed-dose combination.

Target dose and range: A target dose (range) of 4 (2–10) mg/kg bw per day artesunate given once a day for 3 days and a single administration of at least 25 / 1.25 (25–70 / 1.25–3.5) mg/kg bw sulfadoxine / pyrimethamine given as a single dose on day 1.

Body weight (kg)Artesunate dose given daily for 3 days (mg)Sulfadoxine / pyrimethamine dose (mg) given as a single dose on day 1
5 to < 1025 mg250 / 12.5
10 to < 2550 mg500 / 25
25 to < 50100 mg1000 / 50
≥ 50200 mg1500 / 75

Factors associated with altered drug exposure and treatment response: The low dose of folic acid (0.4 mg daily) that is required to protect the fetuses of pregnant women from neural tube defects do not reduce the efficacy of SP, whereas higher doses (5 mg daily) do significantly reduce its efficacy and should not be given concomitantly.

Additional comments:

  • The disadvantage of this ACT is that it is not available as a fixed-dose combination. This may compromise adherence and increase the risk for distribution of loose artesunate tablets, despite the WHO ban on artesunate monotherapy.
  • Resistance is likely to increase with continued widespread use of SP, sulfalene– pyrimethamine and cotrimoxazole (trimethoprim-sulfamethoxazole). Fortunately, molecular markers of resistance to antifols and sulfonamides correlate well with therapeutic responses. These should be monitored in areas in which this drug is used.

Strong recommendation for

Revised dose recommendation for dihydroartemisinin + piperaquine in young children: Children weighing <25kg treated with dihydroartemisinin + piperaquine should receive a minimum of 2.5 mg/kg bw per day of dihydroartemisinin and 20 mg/kg bw per day of piperaquine daily for 3 days.

*unGRADEd recommendation, anticipated to be updated in 2022

Practical Info

Formulations: Currently available as a fixed-dose combination in tablets containing 40 mg dihydroartemisinin and 320 mg piperaquine and paediatric tablets contain 20 mg dihydroartemisinin and 160 mg piperaquine.

Target dose and range: A target dose (range) of 4 (2–10) mg/kg bw per day dihydroartemisinin and 18 (16–27) mg/kg bw per day piperaquine given once a day for 3 days for adults and children weighing ≥ 25 kg. The target doses and ranges for children weighing < 25 kg are 4 (2.5–10) mg/kg bw per day dihydroartemisinin and 24 (20–32) mg/kg bw per day piperaquine once a day for 3 days.

Recommended dosage regimen: The dose regimen currently recommended by the manufacturer provides adequate exposure to piperaquine and excellent cure rates (> 95%), except in children < 5 years, who have a threefold increased risk for treatment failure. Children in this age group have significantly lower plasma piperaquine concentrations than older children and adults given the same mg/kg bw dose. Children weighing < 25 kg should receive at least 2.5 mg/kg bw dihydroartemisinin and 20 mg/kg bw piperaquine to achieve the same exposure as children weighing ≥ 25 kg and adults.

Dihydroartemisinin + piperaquine should be given daily for 3 days.

Body weight (kg)Dihydroartemisinin + piperaquine dose (mg) given daily for 3 days
5 to < 820 + 160
8 to < 1130 + 240
11 to < 1740 + 320
17 to < 2560 + 480
25 to < 3680 + 640
36 to < 60120 + 960
60 < 80160 + 1280
>80200 + 1600

Factors associated with altered drug exposure and treatment response:

High-fat meals should be avoided, as they significantly accelerate the absorption of piperaquine, thereby increasing the risk for potentially arrhythmogenic delayed ventricular repolarization (prolongation of the corrected electrocardiogram QT interval). Normal meals do not alter the absorption of piperaquine.

As malnourished children are at increased risk for treatment failure, their response to treatment should be monitored closely.

  • Dihydroartemisinin exposure is lower in pregnant women.
  • Piperaquine is eliminated more rapidly by pregnant women, shortening the post-treatment prophylactic effect of dihydroartemisinin + piperaquine. As this does not affect primary efficacy, no dosage adjustment is recommended for pregnant women.

Additional comments: Piperaquine prolongs the QT interval by approximately the same amount as chloroquine but by less than quinine. It is not necessary to perform an electrocardiogram before prescribing dihydroartemisinin + piperaquine, but this ACT should not be used in patients with congenital QT prolongation or who have a clinical condition or are on medications that prolong the QT interval. There has been no evidence of cardiotoxicity in large randomized trials or in extensive deployment.

Justification

The dosing subgroup reviewed all available dihydroartemisinin-piperaquine pharmacokinetic data (6 published studies and 10 studies from the WWARN database; total 652 patients) (128)(129) and then conducted simulations of piperaquine exposures for each weight group. These showed lower exposure in younger children with higher risks of treatment failure. The revised dose regimens are predicted to provide equivalent piperaquine exposures across all age groups.

Other considerations

This dose adjustment is not predicted to result in higher peak piperaquine concentrations than in older children and adults, and as there is no evidence of increased toxicity in young children, the GRC concluded that the predicted benefits of improved antimalarial exposure are not at the expense of increased risk.

5.2.4. Recurrent falciparum malaria

Recurrence of P. falciparum malaria can result from re-infection or recrudescence (treatment failure). Treatment failure may result from drug resistance or inadequate exposure to the drug due to sub-optimal dosing, poor adherence, vomiting, unusual pharmacokinetics in an individual, or substandard medicines. It is important to determine from the patient’s history whether he or she vomited the previous treatment or did not complete a full course of treatment.

When possible, treatment failure must be confirmed parasitologically. This may require referring the patient to a facility with microscopy or LDH-based RDTs, as P. falciparum histidine-rich protein-2 (PfHRP2)-based tests may remain positive for weeks after the initial infection, even without recrudescence. Referral may be necessary anyway to obtain second-line treatment. In individual patients, it may not be possible to distinguish recrudescence from re-infection, although lack of resolution of fever and parasitaemia or their recurrence within 4 weeks of treatment are considered failures of treatment with currently recommended ACTs. In many cases, treatment failures are missed because patients are not asked whether they received antimalarial treatment within the preceding 1–2 months. Patients who present with malaria should be asked this question routinely.

Failure within 28 days

The recommended second-line treatment is an alternative ACT known to be effective in the region. Adherence to 7-day treatment regimens (with artesunate or quinine both of which should be co-administered with + tetracycline, or doxycycline or clindamycin) is likely to be poor if treatment is not directly observed; these regimens are no longer generally recommended. The distribution and use of oral artesunate monotherapy outside special centres are strongly discouraged, and quinine-containing regimens are not well tolerated.

Failure after 28 days

Recurrence of fever and parasitaemia > 4 weeks after treatment may be due to either recrudescence or a new infection. The distinction can be made only by PCR genotyping of parasites from the initial and the recurrent infections.

As PCR is not routinely used in patient management, all presumed treatment failures after 4 weeks of initial treatment should, from an operational standpoint, be considered new infections and be treated with the first-line ACT. However, reuse of mefloquine within 60 days of first treatment is associated with an increased risk for neuropsychiatric reactions, and an alternative ACT should be used.

5.2.5. Reducing the transmissibility of treated P. falciparum infections in areas of low-intensity transmission

Strong recommendation for, Low certainty evidence

Reducing the transmissibility of treated P. falciparum infections: In low-transmission areas, give a single dose of 0.25 mg/kg bw primaquine with ACT to patients with P. falciparum malaria (except pregnant women, infants aged < 6 months and women breastfeeding infants aged < 6 months) to reduce transmission. G6PD testing is not required.

Practical Info

In light of concern about the safety of the previously recommended dose of 0.75 mg/kg bw in individuals with G6PD deficiency, a WHO panel reviewed the safety of primaquine as a P. falciparum gametocytocide and concluded that a single dose of 0.25 mg/kg bw of primaquine base is unlikely to cause serious toxicity, even in people with G6PD deficiency (138). Thus, where indicated a single dose of 0.25mg/kg bw of primaquine base should be given on the first day of treatment, in addition to an ACT, to all patients with parasitologically confirmed P. falciparum malaria except for pregnant women, infants < 6 months of age and women breastfeeding infants < 6 months of age, because there are insufficient data on the safety of its use in these groups.

Dosing table based on the most widely currently available tablet strength (7.5mg base)

Body weight (kg)Single dose of primaquine (mg base)
10a to < 25 3.753.75
25 to < 50 7.57.5
50 to 100 1515
a

Dosing of young children weighing < 10 kg is limited by the tablet sizes currently available.

Please refer to the Policy brief on single-dose primaquine as a gametocytocide in Plasmodium falciparum malaria (139).

Evidence To Decision

Benefits and harms

Desirable effects

  • Single doses of primaquine > 0.4 mg/kg bw reduced gametocyte carriage at day 8 by around two thirds (moderate-quality evidence).
  • There are too few trials of doses < 0.4 mg/kg bw to quantify the effect on gametocyte carriage (low-quality evidence).
  • Analysis of observational data from mosquito feeding studies suggests that 0.25 mg/kg bw may rapidly reduce the infectivity of gametocytes to mosquitoes.

Undesirable effects

  • People with severe G6PD deficiency are at risk for haemolysis. At this dose, however, the risk is thought to be small; there are insufficient data to quantify this risk.
Certainty of the Evidence Image ch6if2.jpg

Overall certainty of evidence for all critical outcomes: low.

Preference and values

Justification

GRADE

In an analysis of observational studies of single-dose primaquine, data from mosquito feeding studies on 180 people suggest that adding 0.25 mg/kg primaquine to treatment with an ACT can rapidly reduce the infectivity of gametocytes to mosquitoes.

In a systematic review of eight randomized controlled trials of the efficacy of adding single-dose primaquine to ACTs for reducing the transmission of malaria, in comparison with ACTs alone (136):

  • single doses of > 0.4 mg/kg bw primaquine reduced gametocyte carriage at day 8 by about two thirds (RR, 0.34; 95% CI, 0.19–0.59, two trials, 269 participants, high-certainty evidence); and
  • single doses of primaquine > 0.6 mg/kg bw reduced gametocyte carriage at day 8 by about two thirds (RR, 0.29; 95% CI, 0.22–0.37, seven trials, 1380 participants, high-certainty evidence).

There have been no randomized controlled trials of the effects on the incidence of malaria or on transmission to mosquitos.

Other considerations

The guideline development group considered that the evidence of a dose– response relation from observational studies of mosquito feeding was sufficient to conclude the primaquine dose of 0.25mg/kg bw significantly reduced P. falciparum transmissibility.

The population benefits of reducing malaria transmission with gametocytocidal drugs such as primaquine require that a very high proportion of treated patients receive these medicines and that there is no large transmission reservoir of asymptomatic parasite carriers. This strategy is therefore likely to be effective only in areas of low-intensity malaria transmission, as a component of elimination programmes.

Remarks

This recommendation excludes high-transmission settings, as symptomatic patients make up only a small proportion of the total population carrying gametocytes within a community, and primaquine is unlikely to affect transmission.

A major concern of national policy-makers in using primaquine has been the small risk for haemolytic toxicity in G6PD-deficient people, especially where G6PD testing is not available.

Life-threatening haemolysis is considered unlikely with the 0.25mg/kg bw dose and without G6PD testing (137).

Rationale for the recommendation: The Guideline Development Group considered the evidence on dose–response relations in the observational mosquito-feeding studies of reduced transmissibility with the dose of 0.25 mg/kg bw and the judgement of the WHO Evidence Review Group (November 2012). Their view was that the potential public health benefits of single low-dose (0.25 mg/kg bw) primaquine in addition to an ACT for falciparum malaria, without G6PD testing, outweigh the potential risk for adverse effects.

5.3. Treating special risk groups

Several important patient sub-populations, including young children, pregnant women and patients taking potent enzyme inducers (e.g. rifampicin, efavirenz), have altered pharmacokinetics, resulting in sub-optimal exposure to antimalarial drugs. This increases the rate of treatment failure with current dosage regimens. The rates of treatment failure are substantially higher in hyperparasitaemic patients and patients in areas with artemisinin-resistant falciparum malaria, and these groups require greater exposure to antimalarial drugs (longer duration of therapeutic concentrations) than is achieved with current ACT dosage recommendations. It is often uncertain how best to achieve this. Options include increasing individual doses, changing the frequency or duration of dosing, or adding an additional antimalarial drug. Increasing individual doses may not, however, achieve the desired exposure (e.g., lumefantrine absorption becomes saturated), or the dose may be toxic due to transiently high plasma concentrations (piperaquine, mefloquine, amodiaquine, pyronaridine). An additional advantage of lengthening the duration of treatment (by giving a 5-day regimen) is that it provides additional exposure of the asexual cycle to the artemisinin component as well as augmenting exposure to the partner drug. The acceptability, tolerability, safety and effectiveness of augmented ACT regimens in these special circumstances should be evaluated urgently.

Large and obese adults

Large adults are at risk for under-dosing when they are dosed by age or in standard pre-packaged adult weight-based treatments. In principle, dosing of large adults should be based on achieving the target mg/kg bw dose for each antimalarial regimen. The practical consequence is that two packs of an antimalarial drug might have to be opened to ensure adequate treatment. For obese patients, less drug is often distributed to fat than to other tissues; therefore, they should be dosed on the basis of an estimate of lean body weight, ideal body weight. Patients who are heavy but not obese require the same mg/kg bw doses as lighter patients.

In the past, maximum doses have been recommended, but there is no evidence or justification for this practice. As the evidence for an association between dose, pharmacokinetics and treatment outcome in overweight or large adults is limited, and alternative dosing options have not been assessed in treatment trials, it is recommended that this gap in knowledge be assessed urgently. In the absence of data, treatment providers should attempt to follow up the treatment outcomes of large adults whenever possible.

5.3.1. Pregnant and lactating women

Malaria in pregnancy is associated with low-birth-weight infants, increased anaemia and, in low-transmission areas, increased risks for severe malaria, pregnancy loss and death. In high-transmission settings, despite the adverse effects on fetal growth, malaria is usually asymptomatic in pregnancy or is associated with only mild, non-specific symptoms. There is insufficient information on the safety, efficacy and pharmacokinetics of most antimalarial agents in pregnancy, particularly during the first trimester.

First trimester of pregnancy

See Justification under recommendation.

Second and third trimesters

Experience with artemisinin derivatives in the second and third trimesters (over 4000 documented pregnancies) is increasingly reassuring: no adverse effects on the mother or fetus have been reported. The current assessment of risk–benefit suggests that ACTs should be used to treat uncomplicated falciparum malaria in the second and third trimesters of pregnancy. The current standard six-dose artemether + lumefantrine regimen for the treatment of uncomplicated falciparum malaria has been evaluated in > 1000 women in the second and third trimesters in controlled trials and has been found to be well tolerated and safe. In a low-transmission setting on the Myanmar–Thailand border, however, the efficacy of the standard six-dose artemether + lumefantrine regimen was inferior to 7 days of artesunate monotherapy. The lower efficacy may have been due to lower drug concentrations in pregnancy, as was also recently observed in a high-transmission area in Uganda and the United Republic of Tanzania. Although many women in the second and third trimesters of pregnancy in Africa have been exposed to artemether + lumefantrine, further studies are under way to evaluate its efficacy, pharmacokinetics and safety in pregnant women. Similarly, many pregnant women in Africa have been treated with amodiaquine alone or combined with SP or artesunate; however, amodiaquine use for the treatment of malaria in pregnancy has been formally documented in only > 1300 pregnancies. Use of amodiaquine in women in Ghana in the second and third trimesters of pregnancy was associated with frequent minor side- effects but not with liver toxicity, bone marrow depression or adverse neonatal outcomes.

Dihydroartemisinin + piperaquine was used successfully in the second and third trimesters of pregnancy in > 2000 women on the Myanmar–Thailand border for rescue therapy and in Indonesia for first-line treatment. SP, although considered safe, is not appropriate for use as an artesunate partner drug in many areas because of resistance to SP. If artesunate + SP is used for treatment, co-administration of daily high doses (5 mg) of folate supplementation should be avoided, as this compromises the efficacy of SP. A lower dose of folate (0.4–0.5 mg bw/day) or a treatment other than artesunate + SP should be used.

Mefloquine is considered safe for the treatment of malaria during the second and third trimesters; however, it should be given only in combination with an artemisinin derivative.

Quinine is associated with an increased risk for hypoglycaemia in late pregnancy, and it should be used (with clindamycin) only if effective alternatives are not available.

Primaquine and tetracyclines should not be used in pregnancy.

Dosing in pregnancy

Data on the pharmacokinetics of antimalarial agents used during pregnancy are limited. Those available indicate that pharmacokinetic properties are often altered during pregnancy but that the alterations are insufficient to warrant dose modifications at this time. With quinine, no significant differences in exposure have been seen during pregnancy. Studies of the pharmacokinetics of SP used in IPTp in many sites show significantly decreased exposure to sulfadoxine, but the findings on exposure to pyrimethamine are inconsistent. Therefore, no dose modification is warranted at this time.

Studies are available of the pharmacokinetics of artemether + lumefantrine, artesunate + mefloquine and dihydroartemisinin + piperaquine. Most data exist for artemether + lumefantrine; these suggest decreased overall exposure during the second and third trimesters. Simulations suggest that a standard six-dose regimen of lumefantrine given over 5 days, rather than 3 days, improves exposure, but the data are insufficient to recommend this alternative regimen at present. Limited data on pregnant women treated with dihydroartemesinin + piperaquine suggest lower dihydroartemisinin exposure and no overall difference in total piperaquine exposure, but a shortened piperaquine elimination half-life was noted. The data on artesunate + mefloquine are insufficient to recommend an adjustment of dosage. No data are available on the pharmacokinetics of artesunate + amodiaquine in pregnant women with falciparum malaria, although drug exposure was similar in pregnant and non-pregnant women with vivax malaria.

Lactating women

The amounts of antimalarial drugs that enter breast milk and are consumed by breastfeeding infants are relatively small. Tetracycline is contraindicated in breastfeeding mothers because of its potential effect on infants’ bones and teeth. Pending further information on excretion in breast milk, primaquine should not be used for nursing women, unless the breastfed infant has been checked for G6PD deficiency.

Strong recommendation for

Treat pregnant women with uncomplicated P. falciparum malaria during the first trimester with 7 days of quinine + clindamycin.

*unGRADEd recommendation, anticipated to be updated in 2022

Practical Info

Because organogenesis occurs mainly in the first trimester, this is the time of greatest concern for potential teratogenicity, although development of the nervous system continues throughout pregnancy. The antimalarial medicines considered safe in the first trimester of pregnancy are quinine, chloroquine, clindamycin and proguanil.

The safest treatment regimen for pregnant women in the first trimester with uncomplicated falciparum malaria is therefore quinine + clindamycin (10mg/kg bw twice a day) for 7 days (or quinine monotherapy if clindamycin is not available). An ACT or oral artesunate + clindamycin is an alternative if quinine + clindamycin is not available or fails.

In reality, women often do not declare their pregnancy in the first trimester or may not yet be aware that they are pregnant. Therefore, all women of childbearing age should be asked about the possibility that they are pregnant before they are given antimalarial agents; this is standard practice for administering any medicine to potentially pregnant women. Nevertheless, women in early pregnancy will often be exposed inadvertently to the available first-line treatment, mostly ACT. Published prospective data on 700 women exposed in the first trimester of pregnancy indicate no adverse effects of artemisinins (or the partner drugs) on pregnancy or on the health of fetuses or neonates. The available data are sufficient to exclude a ≥ 4.2-fold increase in risk of any major defect detectable at birth (background prevalence assumed to be 0.9%), if half the exposures occur during the embryo-sensitive period (4–9 weeks post-conception). These data provide assurance in counselling women exposed to an antimalarial drug early in the first trimester and indicate that there is no need for them to have their pregnancy interrupted because of this exposure.

Dosing in pregnancy

Data on the pharmacokinetics of antimalarial agents used during pregnancy are limited. Those available indicate that pharmacokinetic properties are often altered during pregnancy but that the alterations are insufficient to warrant dose modifications at this time. With quinine, no significant differences in exposure have been seen during pregnancy. Studies of the pharmacokinetics of SP used in IPTp in many sites show significantly decreased exposure to sulfadoxine, but the findings on exposure to pyrimethamine are inconsistent. Therefore, no dose modification is warranted at this time.

Studies are available of the pharmacokinetics of artemether + lumefantrine, artesunate + mefloquine and dihydroartemisinin + piperaquine. Most data exist for artemether + lumefantrine; these suggest decreased overall exposure during the second and third trimesters. Simulations suggest that a standard six-dose regimen of lumefantrine given over 5 days, rather than 3 days, improves exposure, but the data are insufficient to recommend this alternative regimen at present. Limited data on pregnant women treated with dihydroartemesinin + piperaquine suggest lower dihydroartemisinin exposure and no overall difference in total piperaquine exposure, but a shortened piperaquine elimination half-life was noted. The data on artesunate + mefloquine are insufficient to recommend an adjustment of dosage. No data are available on the pharmacokinetics of artesunate + amodiaquine in pregnant women with falciparum malaria, although drug exposure was similar in pregnant and non-pregnant women with vivax malaria.

Evidence To Decision

Benefits and harms

Undesirable effects:

  • Published prospective data on 700 women exposed in the first trimester of pregnancy have not indicated any adverse effects of artemisinin-derivatives on pregnancy or on the health of the fetus or neonate.
  • The currently available data are only sufficient to exclude a ≥ 4.2-fold increase in risk of any major defect detectable at birth (background prevalence assumed to be 0.9%), if half the exposures occur during the embryo-sensitive period (4–9 weeks post-conception).

Preference and values

Justification

Evidence supporting the recommendation

Data available were not suitable for evaluation using the GRADE methodology, as there is no /almost no evidence for alternative treatment using ACT.

Safety assessment from published prospective data on 700 women exposed in the first trimester of pregnancy has not indicated any adverse effects of artemisinin-derivatives on pregnancy or on the health of the fetus or neonate.

The currently available data are only sufficient to exclude a ≥ 4.2-fold increase in risk of any major defect detectable at birth (background prevalence assumed to be 0.9%), if half the exposures occur during the embryo-sensitive period (4–9 weeks post-conception).

Other considerations

The limited data available on the safety of artemisinin-derivatives in early pregnancy allow for some reassurance in counselling women accidentally exposed to an artemisinin-derivative early in the first trimester. There is no need for them to have their pregnancy interrupted because of this exposure.

In the absence of adequate safety data on the artemisinin-derivatives in the first trimester of pregnancy the Guideline Development Group was unable to make recommendations beyond reiterating the status quo.

Remarks

Previous data indicated that the antimalarial medicines considered safe in the first trimester of pregnancy are quinine, chloroquine, clindamycin and proguanil. This evidence was not revisited during this guideline process.

The limited data available on the safety of artemisinin-derivatives in early pregnancy allow for some reassurance in counselling women accidentally exposed to an artemisinin-derivative early in the first trimester, and there is no need for them to have their pregnancy interrupted because of this exposure (134)(135).

Rationale for the recommendation

In the absence of adequate safety data on the artemisinin-derivatives in the first trimester of pregnancy the Guideline Development Group was unable to make recommendations beyond reiterating the status quo.

5.3.2. Young children and infants

Artemisinin derivatives are safe and well tolerated by young children; therefore, the choice of ACT is determined largely by the safety and tolerability of the partner drug.

SP (with artesunate) should be avoided in the first weeks of life because it displaces bilirubin competitively and could thus aggravate neonatal hyperbilibinaemia. Primaquine should be avoided in the first 6 months of life (although there are no data on its toxicity in infants), and tetracyclines should be avoided throughout infancy. With these exceptions, none of the other currently recommended antimalarial treatments has shown serious toxicity in infancy.

Delay in treating P. falciparum malaria in infants and young children can have fatal consequences, particularly for more severe infections. The uncertainties noted above should not delay treatment with the most effective drugs available. In treating young children, it is important to ensure accurate dosing and retention of the administered dose, as infants are more likely to vomit or regurgitate antimalarial treatment than older children or adults. Taste, volume, consistency and gastrointestinal tolerability are important determinants of whether the child retains the treatment. Mothers often need advice on techniques of drug administration and the importance of administering the drug again if it is regurgitated within 1 h of administration. Because deterioration in infants can be rapid, the threshold for use of parenteral treatment should be much lower.

Optimal antimalarial dosing in young children

Although dosing on the basis of body area is recommended for many drugs in young children, for the sake of simplicity, antimalarial drugs have been administered as a standard dose per kg bw for all patients, including young children and infants. This approach does not take into account changes in drug disposition that occur with development. The currently recommended doses of lumefantrine, piperaquine, SP, artesunate and chloroquine result in lower drug concentrations in young children and infants than in older patients. Adjustments to previous dosing regimens for dihydroartemisinin + piperaquine in uncomplicated malaria and for artesunate in severe malaria are now recommended to improve the drug exposure in this vulnerable population. The available evidence for artemether + lumefantrine, SP and chloroquine does not indicate dose modification at this time, but young children should be closely monitored, as reduced drug exposure may increase the risk for treatment failure. Limited studies of amodiaquine and mefloquine showed no significant effect of age on plasma concentration profiles.

In community situations where parenteral treatment is needed but cannot be given, such as for infants and young children who vomit antimalarial drugs repeatedly or are too weak to swallow or are very ill, give rectal artesunate and transfer the patient to a facility in which parenteral treatment is possible. Rectal administration of a single dose of artesunate as pre-referral treatment reduces the risks for death and neurological disability, as long as this initial treatment is followed by appropriate parenteral antimalarial treatment in hospital. Further evidence on pre-referral rectal administration of artesunate and other antimalarial drugs is given in section 5.5.3 Treating severe malaria - pre-referral treatment options.

Optimal antimalarial dosing in infants

See recommendation for Infants less than 5 kg body weight below.

Optimal antimalarial dosing in malnourished young children

Malaria and malnutrition frequently coexist. Malnutrition may result in inaccurate dosing when doses are based on age (a dose may be too high for an infant with a low weight for age) or on weight (a dose may be too low for an infant with a low weight for age). Although many studies of the efficacy of antimalarial drugs have been conducted in populations and settings where malnutrition was prevalent, there are few studies of the disposition of the drugs specifically in malnourished individuals, and these seldom distinguished between acute and chronic malnutrition. Oral absorption of drugs may be reduced if there is diarrhoea or vomiting, or rapid gut transit or atrophy of the small bowel mucosa. Absorption of intramuscular and possibly intrarectal drugs may be slower, and diminished muscle mass may make it difficult to administer repeated intramuscular injections to malnourished patients. The volume of distribution of some drugs may be larger and the plasma concentrations lower. Hypoalbuminaemia may reduce protein binding and increase metabolic clearance, but concomitant hepatic dysfunction may reduce the metabolism of some drugs; the net result is uncertain.

Small studies of the pharmacokinetics of quinine and chloroquine showed alterations in people with different degrees of malnutrition. Studies of SP in IPTp and of amodiaquine monotherapy and dihydroartemisinin + piperaquine for treatment suggest reduced efficacy in malnourished children. A pooled analysis of data for individual patients showed that the concentrations of lumefantrine on day 7 were lower in children < 3 years who were underweight for age than in adequately nourished children and adults. Although these findings are concerning, they are insufficient to warrant dose modifications (in mg/kg bw) of any antimalarial drug in patients with malnutrition.

Infants less than 5kg body weight (2015)

Strong recommendation for

Treat infants weighing < 5 kg with uncomplicated P. falciparum malaria with ACT at the same mg/kg bw target dose as for children weighing 5 kg.

*unGRADEd recommendation, anticipated to be updated in 2022

Practical Info

The pharmacokinetics properties of many medicines in infants differ markedly from those in adults because of the physiological changes that occur in the first year of life. Accurate dosing is particularly important for infants. The only antimalarial agent that is currently contraindicated for infants (< 6 months) is primaquine.

ACT is recommended and should be given according to body weight at the same mg/kg bw dose for all infants, including those weighing < 5 kg, with close monitoring of treatment response. The lack of infant formulations of most antimalarial drugs often necessitates division of adult tablets, which can lead to inaccurate dosing. When available, paediatric formulations and strengths are preferred, as they improve the effectiveness and accuracy of ACT dosing.

Evidence To Decision

Benefits and harms

Undesirable effects:

  • There is some evidence that artemether + lumefantrine and dihydroartemisinin + piperaquine may achieve lower plasma concentrations in infants than in older children and adults.

Preference and values

Justification

Evidence supporting the recommendation

Data available were not suitable for evaluation using the GRADE methodology.

In most clinical studies, subgroups of infants and older children were not distinguished, and the evidence for young infants (< 5 kg) is insufficient for confidence in current treatment recommendations. Nevertheless, despite these uncertainties, infants need prompt, effective treatment of malaria. There is limited evidence that artemether + lumefantrine and dihydroartemisinin + piperaquine achieve lower plasma concentrations in infants than in older children and adults.

Other considerations

The Guideline Development Group considered the currently available evidence too limited to warrant formal evidence review at this stage, and was unable to recommend any changes beyond the status quo. Further research is warranted.

Rationale for the recommendation

Treat infants weighing < 5 kg with uncomplicated P. falciparum malaria with an ACT. The weight-adjusted dose should achieve the same mg/kg bw target dose as for children weighing 5 kg.

5.3.3. Patients co-infected with HIV

There is considerable geographical overlap between malaria and HIV infection, and many people are co-infected. Worsening HIV-related immunosuppression may lead to more severe manifestations of malaria. In HIV-infected pregnant women, the adverse effects of placental malaria on birth weight are increased. In areas of stable endemic malaria, HIV-infected patients who are partially immune to malaria may have more frequent, higher-density infections, while in areas of unstable transmission, HIV infection is associated with increased risks for severe malaria and malaria-related deaths. Limited information is available on how HIV infection modifies therapeutic responses to ACTs. Early studies suggested that increasing HIV-related immunosuppression was associated with decreased treatment response to antimalarial drugs.

There is presently insufficient information to modify the general malaria treatment recommendations for patients with HIV/AIDS.

Patients co-infected with tuberculosis

Rifamycins, in particular rifampicin, are potent CYP3A4 inducers with weak antimalarial activity. Concomitant administration of rifampicin during quinine treatment of adults with malaria was associated with a significant decrease in exposure to quinine and a five-fold higher recrudescence rate. Similarly, concomitant rifampicin with mefloquine in healthy adults was associated with a three-fold decrease in exposure to mefloquine. In adults co-infected with HIV and tuberculosis who were being treated with rifampicin, administration of artemether + lumefantrine resulted in significantly lower exposure to artemether, dihydroartemisinin and lumefantrine (nine-, six- and three-fold decreases, respectively). There is insufficient evidence at this time to change the current mg/kg bw dosing recommendations; however, as these patients are at higher risk of recrudescent infections they should be monitored closely.

Patients co-infected with HIV (2015)

Good practice statement

Patients co-infected with HIV: In people who have HIV/AIDS and uncomplicated P. falciparum malaria, avoid artesunate + SP if they are being treated with co-trimoxazole, and avoid artesunate + amodiaquine if they are being treated with efavirenz or zidovudine.

Justification

More data are available on use of artemether + lumefantrine with antiretroviral treatment. A study in children with uncomplicated malaria in a high-transmission area of Africa showed a decreased risk for recurrent malaria after treatment with artemether + lumefantrine in children receiving lopinavir–ritonavir-based antiretroviral treatment as compared with non-nucleoside reverse transcriptase inhibitor-based antiretroviral treatment. Evaluation of pharmacokinetics in these children and in healthy volunteers showed significantly higher exposure to lumefantrine and lower exposure to dihydroartemisinin with lopinavir–ritonavir-based antiretroviral treatment, but no adverse consequences. Conversely, efavirenz-based antiretroviral treatment was associated with a two- to fourfold decrease in exposure to lumefantrine in healthy volunteers and malaria-infected adults and children, with increased rates of recurrent malaria after treatment. Close monitoring is required. Increasing artemether + lumefantrine dosing with efavirenz-based antiretroviral treatment has not yet been studied. Exposure to lumefantrine and other non-nucleoside reverse transcriptase inhibitor-based antiretroviral treatment, namely nevirapine and etravirine, did not show consistent changes that would require dose adjustment.

Studies of administration of quinine with lopinavir–ritonavir or ritonavir alone in healthy volunteers gave conflicting results. The combined data are insufficient to justify dose adjustment. Single-dose atovaquone–proguanil with efavirenz, lopinavir–ritonavir or atazanavir–ritonavir were all associated with a significantly decreased area under the concentration–time curve for atovaquone (two- to fourfold) and proguanil (twofold), which could well compromise treatment or prophylactic efficacy. There is insufficient evidence to change the current mg/kg bw dosing recommendations; however, these patients should also be monitored closely.

5.3.4. Non-immune travellers

Travellers who acquire malaria are often non-immune people living in cities in endemic countries with little or no transmission or are visitors from non-endemic countries travelling to areas with malaria transmission. Both are at higher risk for severe malaria. In a malaria-endemic country, they should be treated according to national policy, provided the treatment recommended has a recent proven cure rate > 90%. Travellers who return to a non-endemic country and then develop malaria present a particular problem, and the case fatality rate is often high; doctors in non-malarious areas may be unfamiliar with malaria and the diagnosis is commonly delayed, and effective antimalarial drugs may not be registered or may be unavailable. However, prevention of transmission or the emergence of resistance are not relevant outside malaria-endemic areas. If the patient has taken chemoprophylaxis, the same medicine should not be used for treatment. Treatment of P. vivax, P. ovale and P. malariae malaria in travellers should be the same as for patients in endemic areas (see section 5.4).

There may be delays in obtaining artesunate, artemether or quinine for the management of severe malaria outside endemic areas. If only parenteral quinidine is available, it should be given, with careful clinical and electrocardiographic monitoring (see section 5.5 Treating severe malaria).

Non-immune travellers (2015)

Strong recommendation for, High certainty evidence

Treat travellers with uncomplicated P. falciparum malaria returning to non-endemic settings with ACT.

Evidence To Decision

Certainty of the Evidence Image ch6if1.jpg
Preference and values

Justification

GRADE

Studies have consistently demonstrated that the five WHO recommended ACTs have less than 5% PCR-adjusted treatment failure rates in settings without resistance to the partner drug (high quality evidence).

Other considerations

The Guideline Development Group considered the evidence of superiority of ACTs over non-ACTs from endemic settings to be equally applicable to those travelling from non-endemic settings.

5.3.5. Uncomplicated hyperparasitaemia

Uncomplicated hyperparasitaemia is present in patients who have ≥ 4% parasitaemia but no signs of severity. They are at increased risk for severe malaria and for treatment failure and are considered an important source of antimalarial drug resistance.

Hyperparasitaemia (2015)

Good practice statement

People with P. falciparum hyperparasitaemia are at increased risk for treatment failure, severe malaria and death and should be closely monitored, in addition to receiving ACT.

Justification

In falciparum malaria, the risk for progression to severe malaria with vital organ dysfunction increases at higher parasite densities. In low-transmission settings, mortality begins to increase when the parasite density exceeds 100 000/µL (~2% parasitaemia). On the north-west border of Thailand, before the general introduction of ACT, parasitaemia > 4% without signs of severity was associated with a 3% mortality rate (about 30-times higher than from uncomplicated falciparum malaria with lower densities) and a six-times higher risk of treatment failure. The relationship between parasitaemia and risks depends on the epidemiological context: in higher-transmission settings, the risk of developing severe malaria in patients with high parasitaemia is lower, but “uncomplicated hyperparasitaemia” is still associated with a significantly higher rate of treatment failure.

Patients with a parasitaemia of 4–10% and no signs of severity also require close monitoring, and, if feasible, admission to hospital. They have high rates of treatment failure. Non-immune people such as travellers and individuals in low-transmission settings with a parasitaemia > 2% are at increased risk and also require close attention. Parasitaemia > 10% is considered to indicate severe malaria in all settings.

It is difficult to make a general recommendation about treatment of uncomplicated hyperparasitaemia, for several reasons: recognizing these patients requires an accurate, quantitative parasite count (they will not be identified from semi-quantitative thick film counts or RDTs), the risks for severe malaria vary considerably, and the risks for treatment failure also vary. Furthermore, little information is available on therapeutic responses in uncomplicated hyperparasitaemia. As the artemisinin component of an ACT is essential in preventing progression to severe malaria, absorption of the first dose must be ensured (atovaquone – proguanil alone should not be used for travellers presenting with uncomplicated hyperparasitaemia). Longer courses of treatment are more effective; both giving longer courses of ACT and preceding the standard 3-day ACT regimen with parenteral or oral artesunate have been used.

5.4. Treating uncomplicated malaria caused by P. vivax, P. ovale, P. malariae or P. knowlesi

Plasmodium vivax accounts for approximately half of all malaria cases outside Africa (3)(142)(143). It is prevalent in the Middle East, Asia, the Western Pacific and Central and South America. With the exception of the Horn, it is rarer in Africa, where there is a high prevalence of the Duffy-negative phenotype, particularly in West Africa, although cases are reported in both Mauritania and Mali (143). In most areas where P. vivax is prevalent, the malaria transmission rates are low (except on the island of New Guinea). Affected populations achieve only partial immunity to this parasite, and so people of all ages are at risk for P. vivax malaria (143). Where both P. falciparum and P. vivax are prevalent, the incidence rates of P. vivax tend to peak at a younger age than for P. falciparum. This is because each P. vivax inoculation may be followed by several relapses. The other human malaria parasite species, P. malariae and P. ovale (which is in fact two sympatric species), are less common. P. knowlesi, a simian parasite, causes occasional cases of malaria in or near forested areas of South-East Asia and the Indian subcontinent (144). In parts of the island of Borneo, P. knowlesi is the predominant cause of human malaria and an important cause of severe malaria

Of the six species of Plasmodium that affect humans, only P. vivax and the two species of P. ovale (145) form hypnozoites, which are dormant parasite stages in the liver that cause relapse weeks to years after the primary infection. P. vivax preferentially invades reticulocytes, and repeated illness causes chronic anaemia, which can be debilitating and sometimes life-threatening, particularly in young children (146). Recurrent vivax malaria is an important impediment to human and economic development in affected populations. In areas where P. falciparum and P. vivax co-exist, intensive malaria control often has a greater effect on P. falciparum, as P. vivax, is more resilient to interventions.

Although P. vivax has been considered to be a benign form of malaria, it may sometimes cause severe disease (147). The major complication is anaemia in young children. In Papua province, Indonesia (147), and in Papua New Guinea (148), where malaria transmission is intense, P. vivax is an important cause of malaria morbidity and mortality, particularly in young infants and children. Occasionally, older patients develop vital organ involvement similar to that in severe and complicated P. falciparum malaria (149)(150). During pregnancy, infection with P. vivax, as with P. falciparum, increases the risk for abortion and reduces birth weight (151)(140). In primigravidae, the reduction in birth weight is approximately two thirds that associated with P. falciparum. In one large series, this effect increased with successive pregnancies (151).

P. knowlesi is a zoonosis that normally affects long- and pig-tailed macaque monkeys. It has a daily asexual cycle, resulting in a rapid replication rate and high parasitaemia. P. knowlesi may cause a fulminant disease similar to severe falciparum malaria (with the exception of coma, which does not occur) (152)(153). Co-infection with other species is common.

Diagnosis

Diagnosis of P. vivax, P. ovale, and P. malariae malaria is based on microscopy. P. knowlesi is frequently misdiagnosed under the microscope, as the young ring forms are similar to those of P. falciparum, the late trophozoites are similar to those of P. malariae, and parasite development is asynchronous. Rapid diagnostic tests based on immunochromatographic methods are available for the detection of P. vivax malaria; however, they are relatively insensitive for detecting P. malariae and P. ovale parasitaemia. Rapid diagnostic antigen tests for human Plasmodium species show poor sensitivity for P. knowlesi infections in humans with low parasitaemia (154).

Treatment

The objectives of treatment of vivax malaria are twofold: to cure the acute blood stage infection and to clear hypnozoites from the liver to prevent future relapses. This is known as “radical cure”.

In areas with chloroquine-sensitive P. vivax

For chloroquine-sensitive vivax malaria, oral chloroquine at a total dose of 25 mg base/kg bw is effective and well tolerated. Lower total doses are not recommended, as these encourage the emergence of resistance. Chloroquine is given at an initial dose of 10 mg base/kg bw, followed by 10 mg/kg bw on the second day and 5 mg/kg bw on the third day. In the past, the initial 10 mg/kg bw dose was followed by 5 mg/kg bw at 6 h, 24 h and 48 h. As residual chloroquine suppresses the first relapse of tropical P. vivax (which emerges about 3 weeks after onset of the primary illness), relapses begin to occur 5–7 weeks after treatment if radical curative treatment with primaquine is not given.

ACTs are highly effective in the treatment of vivax malaria, allowing simplification (unification) of malaria treatment; i.e. all malaria infections can be treated with an ACT. The exception is artesunate + SP, where resistance significantly compromises its efficacy. Although good efficacy of artesunate + SP was reported in one study in Afghanistan, in several other areas (such as South-East Asia) P. vivax has become resistant to SP more rapidly than P. falciparum. The initial response to all ACTs is rapid in vivax malaria, reflecting the high sensitivity to artemisinin derivatives, but, unless primaquine is given, relapses commonly follow. The subsequent recurrence patterns differ, reflecting the elimination kinetics of the partner drugs. Thus, recurrences, presumed to be relapses, occur earlier after artemether + lumefantrine than after dihydroartemisinin + piperaquine or artesunate + mefloquine because lumefantrine is eliminated more rapidly than either mefloquine or piperaquine. A similar temporal pattern of recurrence with each of the drugs is seen in the P. vivax infections that follow up to one third of acute falciparum malaria infections in South-East Asia.

In areas with chloroquine-resistant P. vivax

ACTs containing piperaquine, mefloquine or lumefantrine are the recommended treatment, although artesunate + amodiaquine may also be effective in some areas.

In the systematic review of ACTs for treating P. vivax malaria, dihydroartemisinin + piperaquine provided a longer prophylactic effect than ACTs with shorter half-lives (artemether + lumefantrine, artesunate + amodiaquine), with significantly fewer recurrent parasitaemias during 9 weeks of follow-up (RR, 0.57; 95% CI, 0.40–0.82, three trials, 1066 participants). The half-life of mefloquine is similar to that of piperaquine, but use of dihydroartemisinin + piperaquine in P. vivax mono-infections has not been compared directly in trials with use of artesunate + mefloquine.

Uncomplicated P. ovale, P. malariae or P. knowlesi malaria

Resistance of P. ovale, P. malariae and P. knowlesi to antimalarial drugs is not well characterized, and infections caused by these three species are generally considered to be sensitive to chloroquine. In only one study, conducted in Indonesia, was resistance to chloroquine reported in P. malariae.

The blood stages of P. ovale, P. malariae and P. knowlesi should therefore be treated with the standard regimen of ACT or chloroquine, as for vivax malaria.

Mixed malaria infections

Mixed malaria infections are common in endemic areas. For example, in Thailand, despite low levels of malaria transmission, 8% of patients with acute vivax malaria also have P. falciparum infections, and one third of acute P. falciparum infections are followed by a presumed relapse of vivax malaria (making vivax malaria the most common complication of falciparum malaria).

Mixed infections are best detected by nucleic acid-based amplification techniques, such as PCR; they may be underestimated with routine microscopy. Cryptic P. falciparum infections in vivax malaria can be revealed in approximately 75% of cases by RDTs based on the PfHRP2 antigen, but several RDTs cannot detect mixed infection or have low sensitivity for detecting cryptic vivax malaria. ACTs are effective against all malaria species and so are the treatment of choice for mixed infections.

[3][119][120][120][120][121][122][123][124][124][125][126][127][128]

Blood stage infection (2015)

Good practice statement

If the malaria species is not known with certainty, treat as for uncomplicated.

Strong recommendation for, High certainty evidence

In areas with chloroquine-susceptible infections, treat adults and children with uncomplicated P. vivax, P. ovale, P. malariae or P. knowlesi malaria with either ACT (except pregnant women in their first trimester) or chloroquine.

In areas with chloroquine-resistant infections, treat adults and children with uncomplicated P. vivax, P. ovale, P. malariae or P. knowlesi malaria (except pregnant women in their first trimester) with ACT.

Practical Info

In areas with chloroquine-sensitive P. vivax

For chloroquine-sensitive vivax malaria, oral chloroquine at a total dose of 25 mg base/kg bw is effective and well tolerated. Lower total doses are not recommended, as these encourage the emergence of resistance. Chloroquine is given at an initial dose of 10 mg base/kg bw, followed by 10 mg/kg bw on the second day and 5 mg/kg bw on the third day. In the past, the initial 10-mg/kg bw dose was followed by 5 mg/kg bw at 6 h, 24 h and 48 h. As residual chloroquine suppresses the first relapse of tropical P. vivax (which emerges about 3 weeks after onset of the primary illness), relapses begin to occur 5–7 weeks after treatment if radical curative treatment with primaquine is not given.

ACTs are highly effective in the treatment of vivax malaria, allowing simplification (unification) of malaria treatment; i.e. all malaria infections can be treated with an ACT. The exception is artesunate + SP, where resistance significantly compromises its efficacy. Although good efficacy of artesunate + SP was reported in one study in Afghanistan, in several other areas (such as South-East Asia) P. vivax has become resistant to SP more rapidly than P. falciparum. The initial response to all ACTs is rapid in vivax malaria, reflecting the high sensitivity to artemisinin derivatives, but, unless primaquine is given, relapses commonly follow. The subsequent recurrence patterns differ, reflecting the elimination kinetics of the partner drugs. Thus, recurrences, presumed to be relapses, occur earlier after artemether + lumefantrine than after dihydroartemisinin + piperaquine or artesunate + mefloquine because lumefantrine is eliminated more rapidly than either mefloquine or piperaquine. A similar temporal pattern of recurrence with each of the drugs is seen in the P. vivax infections that follow up to one third of acute falciparum malaria infections in South-East Asia.

In areas with chloroquine-resistant P. vivax

ACTs containing piperaquine, mefloquine or lumefantrine are the recommended treatment, although artesunate + amodiaquine may also be effective in some areas.

In the systematic review of ACTs for treating P. vivax malaria, dihydroartemisinin + piperaquine provided a longer prophylactic effect than ACTs with shorter half-lives (artemether + lumefantrine, artesunate + amodiaquine), with significantly fewer recurrent parasitaemias during 9 weeks of follow-up (RR, 0.57; 95% CI, 0.40–0.82, three trials, 1066 participants). The half-life of mefloquine is similar to that of piperaquine, but use of dihydroartemisinin + piperaquine in P. vivax mono-infections has not been compared directly in trials with use of artesunate + mefloquine.

Uncomplicated P. ovale, P. malariae or P. knowlesi malaria

Resistance of P. ovale, P. malariae and P. knowlesi to antimalarial drugs is not well characterized, and infections caused by these three species are generally considered to be sensitive to chloroquine. In only one study, conducted in Indonesia, was resistance to chloroquine reported in P. malariae.

The blood stages of P. ovale, P. malariae and P. knowlesi should therefore be treated with the standard regimen of ACT or chloroquine, as for vivax malaria.

Mixed Malaria Infections

Mixed malaria infections are common in endemic areas. For example, in Thailand, despite low levels of malaria transmission, 8% of patients with acute vivax malaria also have P. falciparum infections, and one third of acute P. falciparum infections are followed by a presumed relapse of vivax malaria (making vivax malaria the most common complication of falciparum malaria).

Mixed infections are best detected by nucleic acid-based amplification techniques, such as PCR; they may be underestimated with routine microscopy. Cryptic P. falciparum infections in vivax malaria can be revealed in approximately 75% of cases by RDTs based on the PfHRP2 antigen, but several RDTs cannot detect mixed infection or have low sensitivity for detecting cryptic vivax malaria. ACTs are effective against all malaria species and so are the treatment of choice for mixed infections.

Evidence To Decision

Benefits and harms

Desirable effects:

  • ACTs clear parasites more quickly than chloroquine (high-quality evidence).
  • ACTs with long half-lives provide a longer period of suppressive post-treatment prophylaxis against relapses and new infections (high-quality evidence).
  • Simplified national protocols for all forms of uncomplicated malaria.
  • Adequate treatment of undiagnosed P. falciparum in mixed infections.

Certainty of the Evidence Image ch6if1.jpg

Overall certainty of evidence for all critical outcomes: high.

Preference and values

Justification

GRADE

In a systematic review of ACTs for the treatment of P. vivax malaria (149), five trials were conducted in Afghanistan, Cambodia, India, Indonesia and Thailand between 2002 and 2011 with a total of 1622 participants which compared ACTs directly with chloroquine. In comparison with chloroquine:

ACTs cleared parasites from the peripheral blood more quickly (parasitaemia after 24 h of treatment: RR, 0.42; 95% CI, 0.36–0.50, four trials, 1652 participants, high-quality evidence); and

ACTs were at least as effective in preventing recurrent parasitaemia before day 28 (RR, 0.58; 95% CI, 0.18–1.90, five trials, 1622 participants, high-quality evidence).

In four of these trials, few cases of recurrent parasitaemia were seen before day 28 with both chloroquine and ACTs. In the fifth trial, in Thailand in 2011, increased recurrent parasitaemia was seen after treatment with chloroquine (9%), but was infrequent after ACT (2%) (RR, 0.25; 95% CI, 0.09–0.66, one trial, 437 participants).

ACT combinations with long half-lives provided a longer prophylactic effect after treatment, with significantly fewer cases of recurrent parasitaemia between day 28 and day 42 or day 63 (RR, 0.57; 95% CI, 0.40–0.82, three trials, 1066 participants, moderate-quality evidence).

Other considerations

The guideline development group recognized that, in the few settings in which P. vivax is the only endemic species and where chloroquine resistance remains low, the increased cost of ACT may not be worth the small additional benefits. Countries where chloroquine is used for treatment of vivax malaria should monitor for chloroquine resistance and change to ACT when the treatment failure rate is > 10% at day 28.

Remarks

Current methods cannot distinguish recrudescence from relapse or relapse from newly acquired infections, but the aim of treatment is to ensure that the rates of recurrent parasitaemia of any origin are < 10%.

Primaquine has significant asexual stage activity against vivax malaria and augments the therapeutic response to chloroquine. When primaquine is given routinely for 14 days, it may mask low-level chloroquine resistance and prevent vivax recurrence within 28 days.

Rationale for the recommendation

The Guideline Development Group recognized that, in the few settings in which P. vivax is the only endemic species and where chloroquine resistance remains low, the increased cost of ACT may not be worth the small additional benefits. In these settings, chloroquine may still be considered, but countries should monitor chloroquine resistance and change to ACT when the treatment failure rate is > 10% on day 28.

--

Remarks

Current methods do not distinguish recrudescence from relapse or relapse from newly acquired infection, but the aim of treatment is to ensure that the rates of recurrent parasitaemia of any origin is < 10% within 28 days.

When primaquine is not given for radical cure, slowly eliminated ACT that prevents recurrent parasitaemia before day 28 should be used (dihydroartemisinin + piperaquine or artesunate + mefloquine).

Primaquine has significant asexual stage activity against vivax malaria and augments the therapeutic response to chloroquine. When primaquine is given routinely for 14 days, it may mask low-level chloroquine resistance and prevent vivax recurrence within 28 days.

When primaquine is given routinely for 14 days, ACTs with shorter half-lives (artemether + lumefantrine, or artesunate + amodiaquine) may be sufficient to keep the rate of recurrent parasitaemia before day 28 below 10%.

Rationale for the recommendation

The Guideline Development Group recognized that, in the few settings in which P. vivax is the only endemic species and where chloroquine resistance remains low, the increased cost of ACT may not be worth the small additional benefits. In these settings, chloroquine may still be considered, but countries should monitor chloroquine resistance and change to ACT when the treatment failure rate is > 10% on day 28.

Blood stage infection (2015)

Strong recommendation for, Very low certainty evidence

Treat pregnant women in their first trimester who have chloroquine-resistant P. vivax malaria with quinine.

Evidence To Decision

Certainty of the Evidence Image ch6if3.jpg
Preference and values

Justification

In areas with chloroquine-resistant P. vivax

In the first-trimester of pregnancy, quinine should be used in place of ACTs (section 5.3.1).

Good practice statement

The G6PD status of patients should be used to guide administration of primaquine for preventing relapse.

Strong recommendation for, High certainty evidence

To prevent relapse, treat P. vivax or P. ovale malaria in children and adults (except pregnant women, infants aged < 6 months, women breastfeeding infants aged < 6 months, women breastfeeding older infants unless they are known not to be G6PD deficient, and people with G6PD deficiency) with a 14-day course of primaquine in all transmission settings.

Practical Info

Primaquine for preventing relapse

To achieve radical cure (cure and prevention of relapse), relapses originating from liver hypnozoites must be prevented by giving primaquine. The frequency and pattern of relapses varies geographically, with relapse rates generally ranging from 8% to 80%. Temperate long-latency P. vivax strains are still prevalent in many areas. Recent evidence suggests that, in endemic areas where people are inoculated frequently with P. vivax, a significant proportion of the population harbours dormant but “activatable” hypnozoites. The exact mechanism of activation of dormant hypnozoites is unclear. There is evidence that systemic parasitic and bacterial infections, but not viral infections, can activate P. vivax hypnozoites, which explains why P. vivax commonly follows P. falciparum infections in endemic areas where both parasites are prevalent. Thus, the radical curative efficacy of primaquine must be set against the prevalent relapse frequency and the likely burden of “activatable” hypnozoites. Experimental studies on vivax malaria and the relapsing simian malaria P. cynomolgi suggest that the total dose of 8-aminoquinoline given is the main determinant of radical curative efficacy. In most therapeutic assessments, primaquine has been given for 14 days. Total doses of 3.5 mg base/kg bw (0.25 mg/kg bw per day) are required for temperate strains and 7 mg base/kg bw (0.5 mg/kg bw per day) is needed for the tropical, frequent-relapsing P. vivax prevalent in East Asia and Oceania. Primaquine causes dose-limiting abdominal discomfort when taken on an empty stomach; it should always be taken with food.

Use of primaquine to prevent relapse in high-transmission settings was not recommended previously, as the risk for new infections was considered to outweigh any benefits of preventing relapse. This may have been based on underestimates of the morbidity and mortality associated with multiple relapses, particularly in young children. Given the benefits of preventing relapse and in the light of changing epidemiology worldwide and more aggressive targets for malaria control and elimination, the group now recommends that primaquine be used in all settings.

Primaquine formulation: If available, administer scored tablets containing 7.5 or 15 mg of primaquine. Smaller-dose tablets containing 2.5 and 5 mg base are available in some areas and facilitate accurate dosing in children. When scored tablets are not available, 5 mg tablets can be used.

Therapeutic dose: 0.25–0.5 mg/kg bw per day primaquine once a day for 14 days.

Use of primaquine to prevent relapse in high-transmission settings was not recommended previously, as the risk for new infections was considered to outweigh any benefits of preventing relapse. This may have been based on underestimates of the morbidity and mortality associated with multiple relapses, particularly in young children. Given the benefits of preventing relapse and in the light of changing epidemiology worldwide and more aggressive targets for malaria control and elimination, the group now recommends that primaquine be used in all settings.

Evidence To Decision

Benefits and harms

Desirable effects:

  • 14-day courses of primaquine added to chloroquine reduce relapse rates to a greater extent than chloroquine alone (high-quality evidence).
  • 14-day courses of primaquine added to chloroquine may result in fewer relapses than 7-day courses (low-quality evidence).

Undesirable effects:

  • Primaquine is known to cause haemolysis in people with G6PD deficiency.
  • Of the 15 trials included in the Cochrane review, 12 explicitly excluded people with G6PD deficiency; in three trials, it was unclear whether participants were tested for G6PD deficiency or excluded. None of the trials reported serious or treatment-limiting adverse events.

Certainty of the Evidence Image ch6if1.jpg

Overall certainty of evidence for all critical outcomes: high.

Preference and values

Justification

GRADE

In a systematic review of primaquine for radical cure of P. vivax malaria (152), 14 days of primaquine was compared with placebo or no treatment in 10 trials, and 14 days was compared with 7 days in one trial. The trials were conducted in Colombia, Ethiopia, India, Pakistan and Thailand between 1992 and 2006.

In comparison with placebo or no primaquine:

  • 14 days of primaquine (0.25 mg/kg bw per day) reduced relapses during 15 months of follow-up by about 40% (RR, 0.60; 95% CI, 0.48–0.75, 10 trials, 1740 participants, high-quality evidence).

In comparison with 7 days of primaquine:

  • 14 days of primaquine (0.25 mg/kg bw per day) reduced relapses during 6 months of follow-up by over 50% (RR, 0.45; 95% CI, 0.25–0.81, one trial, 126 participants, low-quality evidence).

No direct comparison has been made of higher doses (0.5 mg/kg bw for 14 days) with the standard regimen (0.25 mg/kg bw for 14 days).

Twelve of the 15 trials included in the review explicitly excluded people with G6PD deficiency; the remaining three did not report on this aspect. No serious adverse events were reported.

Other considerations

In the absence of evidence to recommend alternatives, the guideline development group considers 0.75 mg/kg bw primaquine given once weekly for 8 weeks to be the safest regimen for people with mild-to-moderate G6PD deficiency.

Remarks

The widely used primaquine regimen of 0.25 mg base/kg bw per day for 14 days is based on studies of long-latency Korean P. vivax.

In South-East Asia and Oceania, P. vivax relapses at 3-week intervals and is more resistant to primaquine. Consequently, higher doses of primaquine have been used (0.375–0.5 mg base/kg bw per day), but there are few data from comparative trials.

Primaquine is contraindicated in pregnancy and lactation < 6 months post-partum, unless the infant has been tested for G6PD deficiency. It could be given to women who have delivered and ceased breastfeeding.

Rationale for the recommendation

Primaquine has not previously been recommended in high-transmission settings, where the risk of new infections was considered to outweigh any benefits of reduced spontaneous relapses.

In the light of changing epidemiology worldwide and more aggressive targets for malaria control and elimination, the group now recommends primaquine for radical cure of P. vivax in all settings.

Conditional recommendation, Very low certainty evidence

In people with G6PD deficiency, consider preventing relapse by giving primaquine base at 0.75 mg/kg bw once a week for 8 weeks, with close medical supervision for potential primaquine-induced haemolysis.

Practical Info

  • In patients known to be G6PD deficient, primaquine may be considered at a dose of 0.75 mg base/kg bw once a week for 8 weeks. The decision to give or withhold primaquine should depend on the possibility of giving the treatment under close medical supervision, with ready access to health facilities with blood transfusion services.
  • Some heterozygote females who test as normal or not deficient in qualitative G6PD screening tests have intermediate G6PD activity and can still haemolyse substantially. Intermediate deficiency (30–80% of normal) and normal enzyme activity (> 80% of normal) can be differentiated only with a quantitative test. In the absence of quantitative testing, all females should be considered as potentially having intermediate G6PD activity and given the 14-day regimen of primaquine, with counselling on how to recognize symptoms and signs of haemolytic anaemia. They should be advised to stop primaquine and be told where to seek care should these signs develop.
  • If G6PD testing is not available, a decision to prescribe or withhold primaquine should be based on the balance of the probability and benefits of preventing relapse against the risks of primaquine-induced haemolytic anaemia. This depends on the population prevalence of G6PD deficiency, the severity of the prevalent genotypes and on the capacity of health services to identify and manage primaquine-induced haemolytic reactions.

Evidence To Decision

Benefits and harms

Desirable effects:

  • There are no comparative trials of the efficacy or safety of primaquine in people with G6PD deficiency.

Undesirable effects:

  • Primaquine is known to cause haemolysis in people with G6PD deficiency.
  • Of the 15 trials included in the systematic review, 12 explicitly excluded people with G6PD deficiency; in three trials, it was unclear whether participants were tested for G6PD deficiency or excluded. None of the trials reported serious or treatment-limiting adverse events.

Certainty of the Evidence Image ch6if3.jpg

Overall certainty of evidence for all critical outcomes: very low.

Preference and values

Justification

GRADE

In a systematic review of primaquine for radical cure of P. vivax malaria (152), 14 days of primaquine was compared with placebo or no treatment in 10 trials, and 14 days was compared with 7 days in one trial. The trials were conducted in Colombia, Ethiopia, India, Pakistan and Thailand between 1992 and 2006.

In comparison with placebo or no primaquine:

14 days of primaquine (0.25 mg/kg bw per day) reduced relapses during 15 months of follow-up by about 40% (RR, 0.60; 95% CI, 0.48–0.75, 10 trials, 1740 participants, high-quality evidence).

In comparison with 7 days of primaquine:

14 days of primaquine (0.25 mg/kg bw per day) reduced relapses during 6 months of follow-up by over 50% (RR, 0.45; 95% CI, 0.25–0.81, one trial, 126 participants, low-quality evidence).

No direct comparison has been made of higher doses (0.5 mg/kg bw for 14 days) with the standard regimen (0.25 mg/kg bw for 14 days).

Twelve of the 15 trials included in the review explicitly excluded people with G6PD deficiency; the remaining three did not report on this aspect. No serious adverse events were reported.

Other considerations

In the absence of evidence to recommend alternatives, the guideline development group considers 0.75 mg/kg bw primaquine given once weekly for 8 weeks to be the safest regimen for people with mild-to-moderate G6PD deficiency.

Primaquine and glucose-6-phosphate dehydrogenase deficiency

Any person (male or female) with red cell G6PD activity < 30% of the normal mean has G6PD deficiency and will experience haemolysis after primaquine. Heterozygote females with higher mean red cell activities may still show substantial haemolysis. G6PD deficiency is an inherited sex-linked genetic disorder, which is associated with some protection against P. falciparum and P. vivax malaria but increased susceptibility to oxidant haemolysis. The prevalence of G6PD deficiency varies, but in tropical areas it is typically 3–35%; high frequencies are found only in areas where malaria is or has been endemic. There are many (> 180) different G6PD deficiency genetic variants; nearly all of which make the red cells susceptible to oxidant haemolysis, but the severity of haemolysis may vary. Primaquine generates reactive intermediate metabolites that are oxidant and cause variable haemolysis in G6PD-deficient individuals. It also causes methemoglobinaemia. The severity of haemolytic anaemia depends on the dose of primaquine and on the variant of the G6PD enzyme. Fortunately, primaquine is eliminated rapidly so haemolysis is self-limiting once the drug is stopped. In the absence of exposure to primaquine or another oxidant agent, G6PD deficiency rarely causes clinical manifestations so, many patients are unaware of their G6PD status. Screening for G6PD deficiency is not widely available outside hospitals, but rapid screening tests that can be used at points of care have recently become commercially available.

Remarks

Primaquine is contraindicated in pregnancy and lactation, unless the infant has been tested for G6PD deficiency. It could be given to women once they have delivered and ceased breastfeeding.

Rationale for the recommendation

In the absence of evidence to recommend alternatives, the Guideline Development Group considers a regimen of 0.75 mg/kg bw primaquine given once weekly for 8 weeks to be the safest for people with G6PD deficiency.

[139]

Preventing relapse in P. vivax or P. ovale malaria (2015)

Good practice statement

When G6PD status is unknown and G6PD testing is not available, a decision to prescribe primaquine must be based on an assessment of the risks and benefits of adding primaquine.

Justification

If G6PD testing is not available, a decision to prescribe or withhold primaquine should be based on the balance of the probability and benefits of preventing relapse against the risks of primaquine-induced haemolytic anaemia. This depends on the population prevalence of G6PD deficiency, the severity of the prevalent genotypes and on the capacity of health services to identify and manage primaquine-induced haemolytic reactions.

Conditional recommendation, Moderate certainty evidence

Pregnant and breastfeeding women: In women who are pregnant or breastfeeding, consider weekly chemoprophylaxis with chloroquine until delivery and breastfeeding are completed, then, on the basis of G6PD status, treat with primaquine to prevent future relapse.

Practical Info

Primaquine is contraindicated in pregnant women and in lactating women (unless the infant is known not to be G6PD deficient).

As an alternative, chloroquine prophylaxis could be given to suppress relapses after acute vivax malaria during pregnancy.

Once the infant has been delivered and the mother has completed breastfeeding, primaquine could then be given to achieve radical cure.

Few data are available on the safety of primaquine in infancy, and in the past primaquine was not recommended for infants.

There is, however, no specific reason why primaquine should not be given to children aged 6 months to 1 year (provided they do not have G6PD deficiency), as this age group may suffer multiple relapses from vivax malaria. The guideline development group therefore recommended lowering the age restriction to 6 months.

Evidence To Decision

Benefits and harms

Desirable effects:

  • Chloroquine prophylaxis reduced recurrent P. vivax malaria in pregnant women (moderate-quality evidence).

Certainty of the Evidence Image ch6if4.jpg

Overall certainty of evidence for all critical outcomes: moderate.

Preference and values

Justification

GRADE

In a systematic review of malaria chemoprophylaxis in pregnant women (153), chloroquine prophylaxis against P. vivax during pregnancy was directly evaluated in one trial conducted in Thailand in 2001. In comparison with no chemoprophylaxis:

  • Chloroquine prophylaxis substantially reduced recurrent P. vivax malaria (RR, 0.02; 95% CI, 0.00–0.26, one trial, 951 participants, moderate- quality evidence).

Recommendation

Primaquine is contraindicated in pregnant or breastfeeding women with P. vivax malaria. Therefore, consider weekly chemoprophylaxis with chloroquine until delivery and breastfeeding are completed, then treat with 14 days of primaquine to prevent future relapse.

5.5. Treating severe malaria

Mortality from untreated severe malaria (particularly cerebral malaria) approaches 100%. With prompt, effective antimalarial treatment and supportive care, the rate falls to 10–20% overall. Within the broad definition of severe malaria some syndromes are associated with lower mortality rates (e.g. severe anaemia) and others with higher mortality rates (e.g. acidosis). The risk for death increases in the presence of multiple complications.

Any patient with malaria who is unable to take oral medications reliably, shows any evidence of vital organ dysfunction or has a high parasite count is at increased risk for dying. The exact risk depends on the species of infecting malaria parasite, the number of systems affected, the degree of vital organ dysfunction, age, background immunity, pre-morbid, and concomitant diseases, and access to appropriate treatment. Tests such as a parasite count, haematocrit and blood glucose may all be performed immediately at the point of care, but the results of other laboratory measures, if any, may be available only after hours or days. As severe malaria is potentially fatal, any patient considered to be at increased risk should be given the benefit of the highest level of care available. The attending clinician should not worry unduly about definitions: the severely ill patient requires immediate supportive care, and, if severe malaria is a possibility, parenteral antimalarial drug treatment should be started without delay.

Definitions

Severe falciparum malaria: For epidemiological purposes, severe falciparum malaria is defined as one or more of the following, occurring in the absence of an identified alternative cause and in the presence of P. falciparum asexual parasitaemia.

  • Impaired consciousness: A Glasgow coma score < 11 in adults or a Blantyre coma score < 3 in children
  • Prostration: Generalized weakness so that the person is unable to sit, stand or walk without assistance
  • Multiple convulsions: More than two episodes within 24 h
  • Acidosis: A base deficit of > 8 mEq/L or, if not available, a plasma bicarbonate level of < 15 mmol/L or venous plasma lactate ≥ 5 mmol/L. Severe acidosis manifests clinically as respiratory distress (rapid, deep, laboured breathing).
  • Hypoglycaemia: Blood or plasma glucose < 2.2 mmol/L (< 40 mg/dL)
  • Severe malarial anaemia: Haemoglobin concentration ≤ 5 g/dL or a haematocrit of ≤ 15% in children < 12 years of age (< 7 g/dL and < 20%, respectively, in adults) with a parasite count > 10 000/µL
  • Renal impairment: Plasma or serum creatinine > 265 µmol/L (3 mg/dL) or blood urea > 20 mmol/L
  • Jaundice: Plasma or serum bilirubin > 50 µmol/L (3 mg/dL) with a parasite count > 100 000/ µL
  • Pulmonary oedema: Radiologically confirmed or oxygen saturation < 92% on room air with a respiratory rate > 30/min, often with chest indrawing and crepitations on auscultation
  • Significant bleeding: Including recurrent or prolonged bleeding from the nose, gums or venepuncture sites; haematemesis or melaena
  • Shock: Compensated shock is defined as capillary refill ≥ 3 s or temperature gradient on leg (mid to proximal limb), but no hypotension. Decompensated shock is defined as systolic blood pressure < 70 mm Hg in children or < 80 mmHg in adults, with evidence of impaired perfusion (cool peripheries or prolonged capillary refill).
  • Hyperparasitaemia: P. falciparum parasitaemia > 10%

Severe vivax and knowlesi malaria: defined as for falciparum malaria but with no parasite density thresholds.

Severe knowlesi malaria is defined as for falciparum malaria but with two differences:

Therapeutic objectives

The main objective of the treatment of severe malaria is to prevent the patient from dying. Secondary objectives are prevention of disabilities and prevention of recrudescent infection.

Death from severe malaria often occurs within hours of admission to a hospital or clinic, so it is essential that therapeutic concentrations of a highly effective antimalarial drug be achieved as soon as possible. Management of severe malaria comprises mainly clinical assessment of the patient, specific antimalarial treatment, additional treatment and supportive care.

Clinical assessment

Severe malaria is a medical emergency. An open airway should be secured in unconscious patients and breathing and circulation assessed. The patient should be weighed or body weight estimated, so that medicines, including antimalarial drugs and fluids, can be given appropriately. An intravenous cannula should be inserted, and blood glucose (rapid test), haematocrit or haemoglobin, parasitaemia and, in adults, renal function should be measured immediately. A detailed clinical examination should be conducted, including a record of the coma score. Several coma scores have been advocated: the Glasgow coma scale is suitable for adults, and the simple Blantyre modification is easily performed in children. Unconscious patients should undergo a lumbar puncture for cerebrospinal fluid analysis to exclude bacterial meningitis.

The degree of acidosis is an important determinant of outcome; the plasma bicarbonate or venous lactate concentration should be measured, if possible. If facilities are available, arterial or capillary blood pH and gases should be measured in patients who are unconscious, hyperventilating or in shock. Blood should be taken for cross-matching, a full blood count, a platelet count, clotting studies, blood culture and full biochemistry (if possible). Careful attention should be paid to the patient’s fluid balance in severe malaria in order to avoid over- or under-hydration. Individual requirements vary widely and depend on fluid losses before admission.

The differential diagnosis of fever in a severely ill patient is broad. Coma and fever may be due to meningoencephalitis or malaria. Cerebral malaria is not associated with signs of meningeal irritation (neck stiffness, photophobia or Kernig’s sign), but the patient may be opisthotonic. As untreated bacterial meningitis is almost invariably fatal, a diagnostic lumbar puncture should be performed to exclude this condition. There is also considerable clinical overlap between septicaemia, pneumonia and severe malaria, and these conditions may coexist. When possible, blood should always be taken on admission for bacterial culture. In malaria-endemic areas, particularly where parasitaemia is common in young age groups, it is difficult to rule out septicaemia immediately in a shocked or severely ill obtunded child. In all such cases, empirical parenteral broad-spectrum antibiotics should be started immediately, together with antimalarial treatment.

Treatment of severe malaria

It is essential that full doses of effective parenteral (or rectal) antimalarial treatment be given promptly in the initial treatment of severe malaria. This should be followed by a full dose of effective ACT orally. Two classes of medicine are available for parenteral treatment of severe malaria: artemisinin derivatives (artesunate or artemether) and the cinchona alkaloids (quinine and quinidine). Parenteral artesunate is the treatment of choice for all severe malaria. The largest randomized clinical trials ever conducted on severe falciparum malaria showed a substantial reduction in mortality with intravenous or intramuscular artesunate as compared with parenteral quinine. The reduction in mortality was not associated with an increase in neurological sequelae in artesunate-treated survivors. Furthermore, artesunate is simpler and safer to use.

Pre-referral treatment options

See recommendation.

Adjustment of parenteral dosing in renal failure or hepatic dysfunction

The dosage of artemisinin derivatives does not have to be adjusted for patients with vital organ dysfunction. However quinine accumulates in severe vital organ dysfunction. If a patient with severe malaria has persisting acute kidney injury or there is no clinical improvement by 48 h, the dose of quinine should be reduced by one third, to 10 mg salt/kg bw every 12 h. Dosage adjustments are not necessary if patients are receiving either haemodialysis or haemofiltration.

Follow-on treatment

The current recommendation of experts is to give parenteral antimalarial drugs for the treatment of severe malaria for a minimum of 24 h once started (irrespective of the patient’s ability to tolerate oral medication earlier) or until the patient can tolerate oral medication, before giving the oral follow-up treatment.

After initial parenteral treatment, once the patient can tolerate oral therapy, it is essential to continue and complete treatment with an effective oral antimalarial drug by giving a full course of effective ACT (artesunate + amodiaquine, artemether + lumefantrine or dihydroartemisinin + piperaquine). If the patient presented initially with impaired consciousness, ACTs containing mefloquine should be avoided because of an increased incidence of neuropsychiatric complications. When an ACT is not available, artesunate + clindamycin, artesunate + doxycycline, quinine + clindamycin or quinine + doxycycline can be used for follow-on treatment. Doxycycline is preferred to other tetracyclines because it can be given once daily and does not accumulate in cases of renal failure, but it should not be given to children < 8 years or pregnant women. As treatment with doxycycline is begun only when the patient has recovered sufficiently, the 7-day doxycycline course finishes after the artesunate, artemether or quinine course. When available, clindamycin may be substituted in children and pregnant women.

Continuing supportive care

Patients with severe malaria require intensive nursing care, preferably in an intensive care unit where possible. Clinical observations should be made as frequently as possible and should include monitoring of vital signs, coma score and urine output. Blood glucose should be monitored every 4 h, if possible, particularly in unconscious patients.

Management of complications

Severe malaria is associated with a variety of manifestations and complications, which must be recognized promptly and treated as shown below.

Immediate clinical management of severe manifestations and complications of P. falciparum malaria

Manifestation or complicationImmediate managementa
Coma (cerebral malaria)Maintain airway, place patient on his or her side, exclude other treatable causes of coma (e.g. hypoglycaemia, bacterial meningitis); avoid harmful ancillary treatments, intubate if necessary.
HyperpyrexiaAdminister tepid sponging, fanning, a cooling blanket and paracetamol.
ConvulsionsMaintain airways; treat promptly with intravenous or rectal diazepam, lorazepam, midazolam or intramuscular paraldehyde. Check blood glucose.
HypoglycaemiaCheck blood glucose, correct hypoglycaemia and maintain with glucose-containing infusion. Although hypoglycaemia is defined as glucose < 2.2 mmol/L, the threshold for intervention is < 3 mmol/L for children < 5 years and <2.2 mmol/L for older children and adults.
Severe anaemiaTransfuse with screened fresh whole blood.
Acute pulmonary oedemabProp patient up at an angle of 45o, give oxygen, give a diuretic, stop intravenous fluids, intubate and add positive end-expiratory pressure or continuous positive airway pressure in life-threatening hypoxaemia.
Acute kidney injuryExclude pre-renal causes, check fluid balance and urinary sodium; if in established renal failure, add haemofiltration or haemodialysis, or, if not available, peritoneal dialysis.
Spontaneous bleeding and coagulopathyTransfuse with screened fresh whole blood (cryoprecipitate, fresh frozen plasma and platelets, if available); give vitamin K injection.
Metabolic acidosisExclude or treat hypoglycaemia, hypovolaemia and septicaemia. If severe, add haemofiltration or haemodialysis.
ShockSuspect septicaemia, take blood for cultures; give parenteral broad- spectrum antimicrobials, correct haemodynamic disturbances.
a

It is assumed that appropriate antimalarial treatment will have been started in all cases.

b

Prevent by avoiding excess hydration

Additional aspects of management

Fluid therapy

Fluid requirements should be assessed individually. Adults with severe malaria are very vulnerable to fluid overload, while children are more likely to be dehydrated. The fluid regimen must also be adapted to the infusion of antimalarial drugs. Rapid bolus infusion of colloid or crystalloids is contraindicated. If available, haemofiltration should be started early for acute kidney injury or severe metabolic acidosis, which do not respond to rehydration. As the degree of fluid depletion varies considerably in patients with severe malaria, it is not possible to give general recommendations on fluid replacement; each patient must be assessed individually and fluid resuscitation based on the estimated deficit. In high-transmission settings, children commonly present with severe anaemia and hyperventilation (sometimes termed “respiratory distress”) resulting from severe metabolic acidosis and anaemia; they should be treated by blood transfusion. In adults, there is a very thin dividing line between over-hydration, which may produce pulmonary oedema, and under-hydration, which contributes to shock, worsening acidosis and renal impairment. Careful, frequent evaluation of jugular venous pressure, peripheral perfusion, venous filling, skin turgor and urine output should be made.

Blood transfusion

Severe malaria is associated with rapid development of anaemia, as infected, once infected and uninfected erythrocytes are haemolysed and/or removed from the circulation by the spleen. Ideally, fresh, cross-matched blood should be transfused; however, in most settings, cross-matched virus-free blood is in short supply. As for fluid resuscitation, there are not enough studies to make strong evidence-based recommendations on the indications for transfusion; the recommendations given here are based on expert opinion. In high-transmission settings, blood transfusion is generally recommended for children with a haemoglobin level of < 5 g/100 mL (haematocrit < 15%). In low-transmission settings, a threshold of 20% (haemoglobin, 7 g/100 mL) is recommended. These general recommendations must, however, be adapted to the individual, as the pathological consequences of rapid development of anaemia are worse than those of chronic or acute anaemia when there has been adaptation and a compensatory right shift in the oxygen dissociation curve.

Exchange blood transfusion

Many anecdotal reports and several series have claimed the benefit of exchange blood transfusion in severe malaria, but there have been no comparative trials, and there is no consensus on whether it reduces mortality or how it might work. Various rationales have been proposed:

  • removing infected red blood cells from the circulation and therefore lowering the parasite burden (although only the circulating, relatively non-pathogenic stages are removed, and this is also achieved rapidly with artemisinin derivatives);
  • rapidly reducing both the antigen load and the burden of parasite-derived toxins, metabolites and toxic mediators produced by the host; and
  • replacing the rigid unparasitized red cells by more easily deformable cells, therefore alleviating microcirculatory obstruction.

Exchange blood transfusion requires intensive nursing care and a relatively large volume of blood, and it carries significant risks. There is no consensus on the indications, benefits and dangers involved or on practical details such as the volume of blood that should be exchanged. It is, therefore, not possible to make any recommendation regarding the use of exchange blood transfusion.

Concomitant use of antibiotics

The threshold for administering antibiotic treatment should be low in severe malaria. Septicaemia and severe malaria are associated, and there is substantial diagnostic overlap, particularly in children in areas of moderate and high transmission. Thus broad- spectrum antibiotic treatment should be given with antimalarial drugs to all children with suspected severe malaria in areas of moderate and high transmission until a bacterial infection is excluded. After the start of antimalarial treatment, unexplained deterioration may result from a supervening bacterial infection. Enteric bacteria (notably Salmonella) predominated in many trial series in Africa, but a variety of bacteria have been cultured from the blood of patients with a diagnosis of severe malaria.

Patients with secondary pneumonia or with clear evidence of aspiration should be given empirical treatment with an appropriate broad-spectrum antibiotic. In children with persistent fever despite parasite clearance, other possible causes of fever should be excluded, such as systemic Salmonella infections and urinary tract infections, especially in catheterized patients. In the majority of cases of persistent fever, however, no other pathogen is identified after parasite clearance. Antibiotic treatment should be based on culture and sensitivity results or, if not available, local antibiotic sensitivity patterns.

Use of anticonvulsants

The treatment of convulsions in cerebral malaria with intravenous (or, if this is not possible, rectal) benzodiazepines or intramuscular paraldehyde is similar to that for repeated seizures from any cause. In a large, double-blind, placebo-controlled evaluation of a single prophylactic intramuscular injection of 20 mg/kg bw of phenobarbital to children with cerebral malaria, the frequency of seizures was reduced but the mortality rate was increased significantly. This resulted from respiratory arrest and was associated with additional use of benzodiazepine.

A 20 mg/kg bw dose of phenobarbital should not be given without respiratory support. It is not known whether a lower dose would be effective and safer or whether mortality would not increase if ventilation were given. In the absence of further information, prophylactic anticonvulsants are not recommended.

Treatments that are not recommended

In an attempt to reduce the high mortality from severe malaria, various adjunctive treatments have been evaluated, but none has proved effective and many have been shown to be harmful. Heparin, prostacyclin, desferroxamine, pentoxifylline, low-molecular-mass dextran, urea, high-dose corticosteroids, aspirin anti-TNF antibody, cyclosporine A, dichloroacetate, adrenaline, hyperimmune serum,N-acetylcysteine and bolus administration of albumin are not recommended. In addition, use of corticosteroids increases the risk for gastrointestinal bleeding and seizures and has been associated with prolonged coma resolution times when compared with placebo.

Treatment of severe malaria during pregnancy

Women in the second and third trimesters of pregnancy are more likely to have severe malaria than other adults, and, in low-transmission settings, this is often complicated by pulmonary oedema and hypoglycaemia. Maternal mortality is approximately 50%, which is higher than in non-pregnant adults. Fetal death and premature labour are common.

Parenteral antimalarial drugs should be given to pregnant women with severe malaria in full doses without delay. Parenteral artesunate is the treatment of choice in all trimesters. Treatment must not be delayed. If artesunate is unavailable, intramuscular artemether should be given, and if this is unavailable then parenteral quinine should be started immediately until artesunate is obtained.

Obstetric advice should be sought at an early stage, a paediatrician alerted and blood glucose checked frequently. Hypoglycaemia should be expected, and it is often recurrent if the patient is receiving quinine. Severe malaria may also present immediately after delivery. Postpartum bacterial infection is a common complication and should be managed appropriately.

Treatment of severe P. vivax malaria

Although P. vivax malaria is considered to be benign, with a low case-fatality rate, it may cause a debilitating febrile illness with progressive anaemia and can also occasionally cause severe disease, as in P. falciparum malaria. Reported manifestations of severe P. vivax malaria include severe anaemia, thrombocytopenia, acute pulmonary oedema and, less commonly, cerebral malaria, pancytopenia, jaundice, splenic rupture, haemoglobinuria, acute renal failure and shock.

Prompt effective treatment and case management should be the same as for severe P. falciparum malaria (see section 5.5.1). Following parenteral artesunate, treatment can be completed with a full treatment course of oral ACT or chloroquine (in countries where chloroquine is the treatment of choice). A full course of radical treatment with primaquine should be given after recovery.

Please refer to Management of severe malaria - A practical handbook, 3rd edition (160).

5.5.1. Artesunate

Strong recommendation for, High certainty evidence

Treat adults and children with severe malaria (including infants, pregnant women in all trimesters and lactating women) with intravenous or intramuscular artesunate for at least 24 h and until they can tolerate oral medication. Once a patient has received at least 24 h of parenteral therapy and can tolerate oral therapy, complete treatment with 3 days of ACT.

Practical Info

Artesunate is dispensed as a powder of artesunic acid, which is dissolved in sodium bicarbonate (5%) to form sodium artesunate. The solution is then diluted in approximately 5 mL of 5% dextrose and given by intravenous injection or by intramuscular injection into the anterior thigh.

The solution should be prepared freshly for each administration and should not be stored. Artesunate is rapidly hydrolysed in-vivo to dihydroartemisinin, which provides the main antimalarial effect. Studies of the pharmacokinetics of parenteral artesunate in children with severe malaria suggest that they have less exposure than older children and adults to both artesunate and the biologically active metabolite dihydroartemisinin. Body weight has been identified as a significant covariate in studies of the pharmacokinetics of orally and rectally administered artesunate, which suggests that young children have a larger apparent volume of distribution for both compounds and should therefore receive a slightly higher dose of parenteral artesunate to achieve exposure comparable to that of older children and adults.

Artesunate and post-treatment haemolysis

Delayed haemolysis starting >1 week after artesunate treatment of severe malaria has been reported in hyperparasitaemic non-immune travellers. Between 2010 and 2012, there were six reports involving a total of 19 European travellers with severe malaria who were treated with artesunate injection and developed delayed haemolysis. All except one were adults (median age, 50 years; range, 5–71 years). In a prospective study involving African children, the same phenomenon was reported in 5 (7%) of the 72 hyperparasitaemic children studied. Artesunate rapidly kills ring-stage parasites, which are then taken out of the red cells by the spleen; these infected erythrocytes are then returned to the circulation but with a shortened life span, resulting in the observed haemolysis. Thus, post-treatment haemolysis is a predictable event related to the life-saving effect of artesunate. Hyperparasitaemic patients must be followed up carefully to identify late-onset anaemia.

Please refer to the Information note on delayed haemolytic anaemia following treatment with artesunate (156).

Evidence To Decision

Benefits and harms

Desirable effects:

  • In both adults and children, parenteral artesunate prevented more deaths than parenteral quinine (high-quality evidence).
  • For intravenous administration, artesunate is given as a bolus, whereas quinine requires slow infusion.
  • For intramuscular administration, artesunate is given in a smaller volume than quinine.

Undesirable effects:

  • Artesunate is associated with a small increase in neurological sequelae at the time of hospital discharge (moderate-quality evidence). The difference is no longer evident on day 28 after discharge (moderate-quality evidence).

Certainty of the Evidence Image ch6if1.jpg

Overall certainty of evidence for all critical outcomes: high.

Preference and values

Justification

GRADE

In a systematic review of artesunate for severe malaria (155), eight randomized controlled trials with a total of 1664 adults and 5765 children, directly compared parenteral artesunate with parenteral quinine. The trials were conducted in various African and Asian countries between 1989 and 2010.

In comparison with quinine, parenteral artesunate:

  • reduced mortality from severe malaria by about 40% in adults (RR, 0.61; 95% CI, 0.50–0.75, five trials, 1664 participants, high-quality evidence);
  • reduced mortality from severe malaria by about 25% in children (RR, 0.76; 95% CI, 0.65–0.90, four trials, 5765 participants, high-quality evidence); and
  • was associated with a small increase in neurological sequelae in children at the time of hospital discharge (RR, 1.36; 95% CI, 1.01–1.83, three trials, 5163 participants, moderate-quality evidence), most of which, however, slowly resolved, with little or no difference between artesunate and quinine 28 days later (moderate-quality evidence).

Other considerations

The guideline development group considered that the small increase in neurological sequelae at discharge after treatment with artesunate was due to the delayed recovery of the severely ill patients, who would have died had they received quinine. This should not be interpreted as a sign of neurotoxicity. Although the safety of artesunate given in the first trimester of pregnancy has not been firmly established, the guideline development group considered that the proven benefits to the mother outweigh any potential harm to the developing fetus.

Remarks

Parenteral artesunate is recommended as first-line treatment for adults, children, infants and pregnant women in all trimesters of pregnancy.

Rationale for the recommendation

The Guideline Development Group considered the small increase in neurological sequelae at discharge associated with artesunate to be due to prolonged recovery of severely ill patients who would have died if they had received quinine. This should not be interpreted as a sign of neurotoxicity.

Although the safety of artesunate in the first trimester of pregnancy has not been firmly established, the group considered that the proven benefits to the mother outweigh the potential harms to the developing fetus.

Strong recommendation for

Children weighing < 20 kg should receive a higher dose of artesunate (3 mg/kg bw per dose) than larger children and adults (2.4 mg/kg bw per dose) to ensure equivalent exposure to the drug.

*unGRADEd recommendation based on pharmacokinetic modelling, anticipated to be updated in 2022

Practical Info

Artesunate is dispensed as a powder of artesunic acid, which is dissolved in sodium bicarbonate (5%) to form sodium artesunate. The solution is then diluted in approximately 5 mL of 5% dextrose and given by intravenous injection or by intramuscular injection into the anterior thigh.

The solution should be prepared freshly for each administration and should not be stored. Artesunate is rapidly hydrolysed in-vivo to dihydroartemisinin, which provides the main antimalarial effect. Studies of the pharmacokinetics of parenteral artesunate in children with severe malaria suggest that they have less exposure than older children and adults to both artesunate and the biologically active metabolite dihydroartemisinin. Body weight has been identified as a significant covariate in studies of the pharmacokinetics of orally and rectally administered artesunate, which suggests that young children have a larger apparent volume of distribution for both compounds and should therefore receive a slightly higher dose of parenteral artesunate to achieve exposure comparable to that of older children and adults.

Artesunate and post-treatment haemolysis

Delayed haemolysis starting >1 week after artesunate treatment of severe malaria has been reported in hyperparasitaemic non-immune travellers. Between 2010 and 2012, there were six reports involving a total of 19 European travellers with severe malaria who were treated with artesunate injection and developed delayed haemolysis. All except one were adults (median age, 50 years; range, 5–71 years). In a prospective study involving African children, the same phenomenon was reported in 5 (7%) of the 72 hyperparasitaemic children studied. Artesunate rapidly kills ring-stage parasites, which are then taken out of the red cells by the spleen; these infected erythrocytes are then returned to the circulation but with a shortened life span, resulting in the observed haemolysis. Thus, post-treatment haemolysis is a predictable event related to the life-saving effect of artesunate. Hyperparasitaemic patients must be followed up carefully to identify late-onset anaemia.

Justification

The dosing subgroup reviewed all available pharmacokinetic data on artesunate and the main biologically active metabolite dihydroartemisinin following administration of artesunate in severe malaria (published pharmacokinetic studies from 71 adults and 265 children) (157)(158). Simulations of artesunate and dihydroartemisinin exposures were conducted for each age group. These showed underexposure in younger children. The revised parenteral dose regimens are predicted to provide equivalent artesunate and dihydroartemisinin exposures across all age groups.

Other considerations

Individual parenteral artesunate doses between 1.75 and 4 mg/kg have been studied and no toxicity has been observed. The GRC concluded that the predicted benefits of improved antimalarial exposure in children are not at the expense of increased risk.

5.5.2. Parenteral alternatives when artesunate is not available

Conditional recommendation, Low certainty evidence

If artesunate is not available, use artemether in preference to quinine for treating children and adults with severe malaria.

Practical Info

Artemether

Artemether is two to three times less active than its main metabolite dihydroartemisinin. Artemether can be given as an oil-based intramuscular injection or orally. In severe falciparum malaria, the concentration of the parent compound predominates after intramuscular injection, whereas parenteral artesunate is hydrolysed rapidly and almost completely to dihydroartemisinin. Given intramuscularly, artemether may be absorbed more slowly and more erratically than water-soluble artesunate, which is absorbed rapidly and reliably after intramuscular injection. These pharmacological advantages may explain the clinical superiority of parenteral artesunate over artemether in severe malaria.

Artemether is dispensed dissolved in oil (groundnut, sesame seed) and given by intramuscular injection into the anterior thigh.

Therapeutic dose: The initial dose of artemether is 3.2 mg/kg bw intramuscularly (to the anterior thigh). The maintenance dose is 1.6 mg/kg bw intramuscularly daily.

Quinine

Quinine treatment for severe malaria was established before the methods for modern clinical trials were developed. Several salts of quinine have been formulated for parenteral use, but the dihydrochloride is the most widely used. The peak concentrations after intramuscular quinine in severe malaria are similar to those after intravenous infusion. Studies of pharmacokinetics show that a loading dose of quinine (20 mg salt/kg bw, twice the maintenance dose) provides therapeutic plasma concentrations within 4 h. The maintenance dose of quinine (10 mg salt/ kg bw) is administered at 8-h intervals, starting 8 h after the first dose. If there is no improvement in the patient’s condition within 48 h, the dose should be reduced by one third, i.e. to 10 mg salt/kg bw every 12 h.

Rapid intravenous administration of quinine is dangerous. Each dose of parenteral quinine must be administered as a slow, rate-controlled infusion (usually diluted in 5% dextrose and infused over 4 h). The infusion rate should not exceed 5 mg salt/kg bw per h.

Whereas many antimalarial drugs are prescribed in terms of base, for historical reasons quinine doses are usually recommended in terms of salt (usually sulphate for oral use and dihydrochloride for parenteral use). Recommendations for the doses of this and other antimalarial agents should state clearly whether the salt or the base is being referred to; doses with different salts must have the same base equivalents. Quinine must never be given by intravenous bolus injection, as lethal hypotension may result.

Quinine dihydrochloride should be given by rate-controlled infusion in saline or dextrose solution. If this is not possible, it should be given by intramuscular injection to the anterior thigh; quinine should not be injected into the buttock in order to avoid sciatic nerve injury. The first dose should be split, with 10 mg/kg bw into each thigh. Undiluted quinine dihydrochloride at a concentration of 300 mg/ mL is acidic (pH 2) and painful when given by intramuscular injection, so it is best to administer it either in a buffered formulation or diluted to a concentration of 60–100 mg/mL for intramuscular injection. Gluconate salts are less acidic and better tolerated than the dihydrochloride salt when given by the intramuscular and rectal routes.

As the first (loading) dose is the most important in the treatment of severe malaria, it should be reduced only if there is clear evidence of adequate pre-treatment before presentation. Although quinine can cause hypotension if administered rapidly, and overdose is associated with blindness and deafness, these adverse effects are rare in the treatment of severe malaria. The dangers of insufficient treatment (i.e. death from malaria) exceed those of excessive initial treatment.

Evidence To Decision

Benefits and harms

Is parenteral artesunate superior to parenteral quinine in preventing death from severe malaria?

Desirable effects:

  • In children > 12 years and adults, parenteral artesunate probably prevents more deaths than intramuscular artemether (moderate-quality evidence).
  • No randomized controlled trials have been conducted in children aged ≤ 12 years.

--

Is intramuscular artemether superior to parenteral quinine in preventing death from severe malaria?

Desirable effects:

  • In children, artemether is probably equivalent to quinine in preventing death (moderate-quality evidence).
  • In children > 5 years and adults, artemether may be superior to quinine (moderate-quality evidence).
  • Artemether is easier to administer, requiring a smaller fluid volume for intramuscular injection.

Certainty of the Evidence Image ch6if2.jpg

Is parenteral artesunate superior to parenteral quinine in preventing death from severe malaria?

Overall certainty of evidence for all critical outcomes: moderate.

--

Is intramuscular artemether superior to parenteral quinine in preventing death from severe malaria?

Overall certainty of evidence for all critical outcomes: moderate.

Preference and values

Justification

GRADE

A systematic review of intramuscular artemether for severe malaria comprised two randomized controlled trials in Viet Nam in which artemether was compared with artesunate in 494 adults, and 16 trials in Africa and Asia in which artemether was compared with quinine in 716 adults and 1447 children (159). The trials were conducted between 1991 and 2009.

In comparison with artesunate, intramuscular artemether was not as effective at preventing deaths in adults in Asia (RR, 1.80; 95% CI, 1.09–2.97; two trials, 494 participants, moderate-quality evidence).

Artemether and artesunate have not been directly compared in randomized trials in African children.

In comparison with quinine:

  • Intramuscular artemether prevented a similar number of deaths in children in Africa (RR, 0.96; 95% CI, 0.76–1.20; 12 trials, 1447 participants, moderate-quality evidence).
  • Intramuscular artemether prevented more deaths in adults in Asia (RR, 0.59; 95% CI, 0.42–0.83; four trials, 716 participants, moderate-quality evidence).

Other considerations

Indirect comparisons of parenteral artesunate and quinine and of artemether and quinine were considered by the guideline development group with what is known about the pharmacokinetics of the two drugs. They judged the accumulated indirect evidence to be sufficient to recommend parenteral artesunate rather than intramuscular artemether for use in all age groups.

--

Is parenteral artesunate superior to parenteral quinine in preventing death from severe malaria?

Remarks

Intramuscular artemether should be considered only when parenteral artesunate is not available.

Recommendation

Treat children and adults with severe malaria with parenteral artesunate for at least 24 h.

Strength of recommendation: Strong for.

Rationale for the recommendation

Indirect comparisons of artesunate and quinine and of artemether and quinine were considered by the Guideline Development Group, with what is known about the pharmacokinetics of the two drugs. The group considered that the accumulated indirect evidence is sufficient to recommend artesunate over artemether for all age groups.

--

Is intramuscular artemether superior to parenteral quinine in preventing death from severe malaria?

Remarks

Quinine is retained as an option for treating severe malaria when artesunate or artemether is not available or is contraindicated.

Recommendation

If parenteral artesunate is not available, use artemether in preference to quinine for treating children and adults with severe malaria.

Strength of recommendation: conditional for.

Rationale for the recommendation

The Guideline Development Group considered the possible superiority, the ease of administration and the better adverse-event profile of artemether as sufficient to recommend artemether over quinine as a second-line treatment option for severe malaria.

5.5.3. Pre-referral treatment options

The risk for death from severe malaria is greatest in the first 24 h, yet, in most malaria-endemic countries, the transit time between referral and arrival at a health facility where intravenous treatment can be administered is usually long, thus delaying the start of appropriate antimalarial treatment. During this time, the patient may deteriorate or die. It is therefore recommended that patients, particularly young children, be treated with a first dose of one of the recommended treatments before referral (unless the referral time is <6 h).

The recommended pre-referral treatment options for children <6 years, in descending order of preference, are intramuscular artesunate; rectal artesunate; intramuscular artemether; and intramuscular quinine. For older children and adults, the recommended pre-referral treatment options, in descending order of preference, are intramuscular injections of artesunate; artemether; and quinine.

Administration of an artemisinin derivative by the rectal route as pre-referral treatment is feasible and acceptable even at community level. The only trial of rectal artesunate as pre-referral treatment showed the expected reduction in mortality of young children but unexpectedly found increased mortality in older children and adults. As a consequence, rectal artesunate is recommended for use only in children aged <6 years and only when intramuscular artesunate is not available.

When rectal artesunate is used, patients should be transported immediately to a higher-level facility where intramuscular or intravenous treatment is available. If referral is impossible, rectal treatment could be continued until the patient can tolerate oral medication. At this point, a full course of the recommended ACT for uncomplicated malaria should be administered.

The single dose of 10 mg/kg bw of artesunate when given as a suppository should be administered rectally as soon as a presumptive diagnosis of severe malaria is made. If the suppository is expelled from the rectum within 30 min of insertion, a second suppository should be inserted and the buttocks held together for 10 min to ensure retention of the dose.

Strong recommendation for, Moderate certainty evidence

Where complete treatment of severe malaria is not possible, but injections are available, give adults and children a single intramuscular dose of artesunate, and refer to an appropriate facility for further care. Where intramuscular artesunate is not available use intramuscular artemether or, if that is not available, use intramuscular quinine.

Where intramuscular injection of artesunate is not available, treat children < 6 years with a single rectal dose (10mg/kg bw) of artesunate, and refer immediately to an appropriate facility for further care. Do not use rectal artesunate in older children and adults.

Practical Info

Adjustment of parenteral dosing in renal failure of hepatic dysfunction

The dosage of artemisinin derivatives does not have to be adjusted for patients with vital organ dysfunction. However, quinine accumulates in severe vital organ dysfunction. If a patient with severe malaria has persisting acute kidney injury or there is no clinical improvement by 48 h, the dose of quinine should be reduced by one third, to 10 mg salt/kg bw every 12 h. Dosage adjustments are not necessary if patients are receiving either haemodialysis or haemofiltration.

Follow-on treatment

The current recommendation of experts is to give parenteral antimalarial drugs for the treatment of severe malaria for a minimum of 24 h ounce started (irrespective of the patient’s ability to tolerate oral medication earlier) or until the patient can tolerate oral medication, before giving the oral follow-up treatment.

After initial parenteral treatment, once the patient can tolerate oral therapy, it is essential to continue and complete treatment with an effective oral antimalarial drug by giving a full course of effective ACT (artesunate + amodiaquine, artemether + lumefantrine or dihydroartemisinin + piperaquine). If the patient presented initially with impaired consciousness, ACTs containing mefloquine should be avoided because of an increased incidence of neuropsychiatric complications. When an ACT is not available, artesunate + clindamycin, artesunate + doxycycline, quinine + clindamycin or quinine + doxycycline can be used for follow-on treatment. Doxycycline is preferred to other tetracyclines because it can be given once daily and does not accumulate in cases of renal failure, but it should not be given to children < 8 years or pregnant women. As treatment with doxycycline is begun only when the patient has recovered sufficiently, the 7-day doxycycline course finishes after the artesunate, artemether or quinine course. When available, clindamycin may be substituted in children and pregnant women.

Continuing supportive care

Patients with severe malaria require intensive nursing care, preferably in an intensive care unit where possible. Clinical observations should be made as frequently as possible and should include monitoring of vital signs, coma score and urine output. Blood glucose should be monitored every 4 h, if possible, particularly in unconscious patients.

Please refer to Rectal artesunate for pre-referral treatment of severe malaria (161).

Evidence To Decision

Benefits and harms

Desirable effects:

  • No studies of direct comparison of rectal artesunate with parenteral antimalarial drugs for pre-referral treatment.
  • In hospital care, parenteral artesunate reduces the number of deaths to a greater extent than parenteral quinine (high-quality evidence) and probably reduces the number of deaths from that with intramuscular artemether (moderate-quality evidence).

Certainty of the Evidence Image ch6if4.jpg

Overall certainty of evidence for all critical outcomes: moderate.

Preference and values

Justification

GRADE

In a systematic review of pre-referral treatment for suspected severe malaria, in a single large randomized controlled trial of 17 826 children and adults in Bangladesh, Ghana and the United Republic of Tanzania, pre-referral rectal artesunate was compared with placebo (160).

In comparison with placebo:

  • Rectal artesunate reduced mortality by about 25% in children < 6 years (RR, 0.74; 95% CI, 0.59–0.93; one trial, 8050 participants, moderate- quality evidence).
  • Rectal artesunate was associated with more deaths in older children and adults (RR, 2.21; 95% CI, 1.18–4.15; one trial 4018 participants, low- quality evidence).

Other considerations

The guideline development group could find no plausible explanation for the finding of increased mortality among older children and adults in Asia who received rectal artesunate, which may be due to chance. Further trials would provide clarification but are unlikely to be done. The group was therefore unable to recommend its use in older children and adults.

In the absence of direct evaluations of parenteral antimalarial drugs for pre- referral treatment, the guideline development group considered the known benefits of artesunate in hospitalized patients and downgraded the quality of evidence for pre-referral situations. When intramuscular injections can be given, the group recommends intramuscular artesunate in preference to rectal artesunate.

Remarks

This recommendation applies to all people with suspected severe malaria, including infants, lactating women and pregnant women in all trimesters.

Where intramuscular artesunate is not available, use rectal artesunate (in children < 6 years), intramuscular artemether or intramuscular quinine.

Rationale for the recommendation

In the absence of direct comparative evaluations of parenteral antimalarial drugs for pre-referral treatment, the Guideline Development Group considered the known benefits of artesunate in hospitalized patients and downgraded the quality of evidence for use in pre-referral situations. When intramuscular injections can be given, the panel recommends intramuscular artesunate in preference to rectal artesunate.

5.6. Other considerations in treating malaria

5.6.1. Management of malaria cases in special situations

Epidemics and humanitarian emergencies

Environmental, political and economic changes, population movement and war can all contribute to the emergence or re-emergence of malaria in areas where it was previously eliminated or well controlled. The displacement of large numbers of people with little or no immunity within malaria-endemic areas increases the risk for malaria epidemics among the displaced population, while displacement of people from an endemic area to an area where malaria has been eliminated can result in re-introduction of transmission and a risk for epidemics in the resident population.

Climate change may also alter transmission patterns and the malaria burden globally by producing conditions that favour vector breeding and thereby increasing the risks for malaria transmission and epidemics.

Parasitological diagnosis during epidemics

In the acute phase of epidemics and complex emergency situations, facilities for laboratory diagnosis with good-quality equipment and reagents and skilled technicians are often not available or are overwhelmed. Attempts should be made to improve diagnostic capacity rapidly, including provision of RDTs. If diagnostic testing is not feasible, the most practical approach is to treat all febrile patients as suspected malaria cases, with the inevitable consequences of over-treatment of malaria and potentially poor management of other febrile conditions. If this approach is used, it is imperative to monitor intermittently the prevalence of malaria as a true cause of fever and revise the policy appropriately. This approach has sometimes been termed “mass fever treatment”. This is not the same as and should not be confused with “mass drug administration”, which is administration of a complete treatment course of antimalarial medicines to every individual in a geographically defined area without testing for infection and regardless of the presence of symptoms.

Management of uncomplicated falciparum malaria during epidemics

The principles of treatment of uncomplicated malaria are the same as those outlined in section 5.2. Active case detection should be undertaken to ensure that as many patients as possible receive adequate treatment, rather than relying on patients to come to a clinic.

Epidemics of mixed falciparum and vivax or vivax malaria

ACTs (except artesunate + SP) should be used to treat uncomplicated malaria in mixed-infection epidemics, as they are highly effective against all malaria species. In areas with pure P. vivax epidemics, ACTs or chloroquine (if prevalent strains are sensitive) should be used.

Anti-relapse therapy for P. vivax malaria

Administration of 14-day primaquine anti-relapse therapy for vivax malaria may be impractical in epidemic situations because of the duration of treatment and the difficulty of ensuring adherence. If adequate records are kept, therapy can be given in the post-epidemic period to patients who have been treated with blood schizontocides.

Malaria elimination settings

Use of gametocytocidal drugs to reduce transmission

ACT reduces P. falciparum gametocyte carriage and transmission markedly, but this effect is incomplete, and patients presenting with gametocytaemia may be infectious for days or occasionally weeks, despite ACT. The strategy of using a single dose of primaquine to reduce infectivity and thus P. falciparum transmission has been widely used in low transmission settings.

Use of primaquine as a P. falciparum gametocytocide has a particular role in programmes to eliminate P. falciparum The population benefits of reducing malaria transmission by gametocytocidal drugs require that a high proportion of patients receive these medicines. WHO recommends the addition of a single dose of primaquine (0.25 mg base/kg bw) to ACT for uncomplicated falciparum malaria as a gametocytocidal medicine, particularly as a component of elimination programmes. A recent review of the evidence on the safety and effectiveness of primaquine as a gametocytocide of P. falciparum indicates that a single dose of 0.25 mg base/kg bw is effective in blocking infectivity to mosquitos and is unlikely to cause serious toxicity in people with any of the G6PD variants. Thus, the G6PD status of the patient does not have to be known before primaquine is used for this indication.

Artemisinin-resistant falciparum malaria

Artemisinin resistance in P. falciparum is now prevalent in parts of Cambodia, the Lao People’s Democratic Republic, Myanmar, Thailand and Viet Nam. There is currently no evidence for artemisinin resistance outside these areas. The particular advantage of artemisinins over other antimalarial drugs is that they kill circulating ring-stage parasites and thus accelerate therapeutic responses. This is lost in resistance to artemisinin. As a consequence, parasite clearance is slowed, and ACT failure rates and gametocytaemia both increase. The reduced efficacy of artemisinin places greater selective pressure on the partner drugs, to which resistance is also increasing. This situation poses a grave threat. In the past chloroquine resistant parasites emerged near the Cambodia–Thailand border and then spread throughout Asia and Africa at a cost of millions of lives. In Cambodia, where artemisinin resistance is worst, none of the currently recommended treatment regimens provides acceptable cure rates (> 90%), and continued use of ineffective drug regimens fuels the spread of resistance. In Cambodia use of atovaquone–proguanil instead of ACT resulted in very rapid emergence of resistance to atovaquone.

In this dangerous, rapidly changing situation, local treatment guidelines cannot be based on a solid evidence base; however, the risks associated with continued use of ineffective regimens are likely to exceed the risks of new, untried regimens with generally safe antimalarial drugs. At the current levels of resistance, the artemisinin derivatives still provide significant antimalarial activity; therefore, longer courses of treatment with existing or new augmented combinations or treatment with new partner medicines (e.g. artesunate + pyronaridine) may be effective. Studies to determine the best treatments for artemisinin-resistant malaria are needed urgently.

It is strongly recommended that single-dose primaquine (as a gametocytocide) be added to all falciparum malaria treatment regimens as described in section 5.2.5. For the treatment of severe malaria in areas with established artemisinin resistance, it is recommended that parenteral artesunate and parenteral quinine be given together in full doses, as described in section 5.5.

5.6.2. Quality of antimalarial drugs

The two general classes of poor-quality medicines are those that are falsified (counterfeit), in which there is criminal intent to deceive and the drug contains little or no active ingredient (and often other potentially harmful substances), and those that are substandard, in which a legitimate producer has included incorrect amounts of active drug and/or excipients in the medicine, or the medicine has been stored incorrectly or for too long and has degraded. Falsified antimalarial tablets and ampoules containing little or no active pharmaceutical ingredients are a major problem in some areas. They may be impossible to distinguish at points of care from the genuine product and may lead to under-dosage and high levels of treatment failure, giving a mistaken impression of resistance, or encourage the development of resistance by providing sub-therapeutic blood levels. They may also contain toxic ingredients.

Substandard drugs result from poor-quality manufacture and formulation, chemical instability or improper or prolonged storage. Artemisinin and its derivatives in particular have built-in chemical instability, which is necessary for their biological action but which causes pharmaceutical problems both in their manufacture and in their co-formulation with other compounds. The problems of instability are accelerated under tropical conditions. The requirement for stringent quality standards is particularly important for this class of compounds. Many antimalarial drugs are stored in conditions of high heat and humidity and sold beyond their expiry dates.

In many malaria-endemic areas, a large proportion of the antimalarial drugs used are generic products purchased in the private sector. They may contain the correct amounts of antimalarial drug, but, because of their formulation, are inadequately absorbed. Antimalarial medicines must be manufactured according to good manufacturing practice, have the correct drug and excipient contents, be proved to have bioavailability that is similar to that of the reference product, have been stored under appropriate conditions and be dispensed before their expiry date.

Tools to assess drug quality at points of sale are being developed, but the capacity of medicines regulatory agencies in most countries to monitor drug quality is still limited. Legal and regulatory frameworks must be strengthened, and there should be greater collaboration between law enforcement agencies, customs and excise authorities and medicines regulatory agencies to deal more effectively with falsified medicines. Private sector drug distribution outlets should have more information and active engagement with regulatory agencies. WHO, in collaboration with other United Nations agencies, has established an international mechanism to prequalify manufacturers of ACTs on the basis of their compliance with internationally recommended standards of manufacture and quality. Manufacturers of antimalarial medicines with prequalified status are listed on the prequalification web site (168).

Antimalarial drug quality (2015)

Good practice statement

National drug and regulatory authorities should ensure that the antimalarial medicines provided in both the public and the private sectors are of acceptable quality, through regulation, inspection and law enforcement.

5.6.3. Monitoring efficacy and safety of antimalarial drugs and resistance

When adapting and implementing these guidelines, countries should also strengthen their systems for monitoring and evaluating their national programmes. The systems should allow countries to track the implementation and impact of new recommendations, better target their programmes to the areas and populations at greatest need and detect decreasing antimalarial efficacy and drug resistance as early as possible.

Routine surveillance

WHO promotes universal coverage with diagnostic testing and antimalarial treatment and strengthened malaria surveillance systems. In the “test, track, treat” initiative, it is recommended that every suspected malaria case is tested, that every confirmed case is treated with a quality-assured antimalarial medicine and that the disease is tracked by timely, accurate surveillance systems. Surveillance and treatment based on confirmed malaria cases will lead to better understanding of the disease burden and enable national malaria control programmes to direct better their resources to where they are most needed.

Therapeutic efficacy

Monitoring of therapeutic efficacy in falciparum malaria involves assessing clinical and parasitological outcomes of treatment for at least 28 days after the start of adequate treatment and monitoring for the reappearance of parasites in blood. The exact duration of post-treatment follow-up is based on the elimination half- life of the partner drug in the ACT being evaluated. Tools for monitoring antimalaria drug efficacy can be found on the WHO website (169).

PCR genotyping should be used in therapeutic monitoring of antimalarial drug efficacy against P. falciparum to distinguish between recrudescence (true treatment failure) and new infections.

An antimalarial medicine that is recommended in the national malaria treatment policy should be changed if the total treatment failure proportion is ≥ 10%, as assessed in vivo by monitoring therapeutic efficacy. A significantly declining trend in treatment efficacy over time, even if failure rates have not yet fallen to the ≥ 10% cut-off, should alert programmes to undertake more frequent monitoring and to prepare for a potential policy change.

Resistance

Antimalarial drug resistance is the ability of a parasite strain to survive and/or multiply despite administration and absorption of an antimalarial drug given in doses equal to or higher than those usually recommended, provided that drug exposure is adequate. Resistance to antimalarial drugs arises because of selection of parasites with genetic changes (mutations or gene amplifications) that confer reduced susceptibility. Resistance has been documented to all classes of antimalarial medicines, including the artemisinin derivatives, and it is a major threat to malaria control.

Widespread inappropriate use of antimalarial drugs exerts a strong selective pressure on malaria parasites to develop high levels of resistance. Resistance can be prevented, or its onset slowed considerably by combining antimalarial drugs with different mechanisms of action and ensuring high cure rates through full adherence to correct dose regimens. If different drugs with different mechanisms of resistance are used together, the emergence and spread of resistance should be slowed.

Clinical and parasitological assessment of therapeutic efficacy should include:

  • confirmation of the quality of the antimalarial medicines tested;
  • molecular genotyping to distinguish between re-infections and recrudescence and to identify genetic markers of drug resistance;
  • studies of parasite susceptibility to antimalarial drugs in culture; and
  • measurement of antimalarial drug levels to assess exposure in cases of slow therapeutic response or treatment failure

Pharmacovigilance

Governments should have effective pharmacovigilance systems (such as the WHO pregnancy registry) to monitor the safety of all drugs, including antimalarial medicines. The safety profiles of the currently recommended antimalarial drugs are reasonably well described and supported by an evidence base of several thousand participants (mainly from clinical trials); however, rare but serious adverse drug reactions will not be detected in clinical trials of this size, particularly if they occur primarily in young children, pregnant women or people with concurrent illness, who are usually under-represented in clinical trials. Rare but serious adverse drug reactions are therefore detected only in prospective phase IV post-marketing studies or population-based pharmacovigilance systems. In particular, more data are urgently needed on the safety of ACTs during the first trimester of pregnancy and on potential interactions between antimalarial and other commonly used medicines.

Good practice statement

All malaria programmes should regularly monitor the therapeutic efficacy of antimalarial drugs using the standard WHO protocols.

Practical Info

Routine monitoring of antimalarial drug efficacy is necessary to ensure effective case management and for early detection of resistance. WHO recommends that the efficacy of first- and second-line antimalarial treatments be tested at least once every 24 months at all sentinel sites. Data collected from studies conducted according to the standard protocol inform national treatment policies.

Please refer to the tools for monitoring antimalarial drug efficacy (163) and Methods for surveillance of antimalarial drug efficacy (164) which includes tools and materials to conduct routine therapeutic efficacy studies (TES). It is a reference for national programmes and investigators conducting routine surveillance studies to assess the efficacy of medicines that have already been registered.

Additional references include:

5.7. National adaptation and implementation

These guidelines provide a generic framework for malaria diagnosis and treatment policies worldwide; however, national policy-makers will be required to adapt these recommendations on the basis of local priorities, malaria epidemiology, parasite resistance and national resources.

National decision-making

National decision-makers are encouraged to adopt inclusive, transparent, rigorous approaches. Broad, inclusive stakeholder engagement in the design and implementation of national malaria control programmes will help to ensure they are feasible, appropriate, equitable and acceptable. Transparency and freedom from financial conflicts of interest will reduce mistrust and conflict, while rigorous evidence-based processes will ensure that the best possible decisions are made for the population.

Information required for national decision-making

Selection of first- and second-line antimalarial medicines will require reliable national data on their efficacy and parasite resistance, which in turn require that appropriate surveillance and monitoring systems are in place (see Monitoring efficacy and safety of antimalaria drugs). In some countries, the group adapting the guidelines for national use might have to re-evaluate the global evidence base with respect to their own context. The GRADE tables may serve as a starting-point for this assessment. Decisions about coverage, feasibility, acceptability and cost may require input from various health professionals, community representatives, health economists, academics and health system managers.

Opportunities and risks

The recommendations made in these guidelines provide an opportunity to improve malaria case management further, to reduce unnecessary morbidity and mortality and to contribute to continued efforts towards elimination. Failure to implement the basic principles of combination therapy and rational use of antimalarial medicines will risk promoting the emergence and spread of drug resistance, which could undo all the recent gains in malaria control and elimination.

General guiding principles for choosing a case management strategy and tools

Choosing a diagnostic strategy

The two methods currently considered suitable for routine patient management are light microscopy and RDTs. Different strategies may be adopted in different health care settings. The choice between RDTs and microscopy depends on local circumstances, including the skills available, the patient case-load, the epidemiology of malaria and use of microscopy for the diagnosis of other diseases. When the case-load of patients with fever is high, the cost of each microscopy test is likely to be less than that of an RDT; however, high-throughput, high-quality microscopy may be less operationally feasible. Although several RDTs allow diagnosis of both P. falciparum and P. vivax infections, microscopy has further advantages, including accurate parasite counting (and thus identification of high parasite density), prognostication in severe malaria, speciation of other malaria parasites and sequential assessment of the response to antimalarial treatment. Microscopy may help to identify other causes of fever. High-quality light microscopy requires well-trained, skilled staff, good staining reagents, clean slides and, often, electricity to power the microscope. It requires a quality assurance system, which is often not well implemented in malaria-endemic countries.

In many areas, malaria patients are treated outside the formal health services, e.g. in the community, at home or by private providers. Microscopy is generally not feasible in the community, but RDTs might be available, allowing access to confirmatory diagnosis of malaria and the correct management of febrile illnesses. The average sensitivity of HRP2-detecting RDTs is generally greater than that of RDTs for detecting pLDH of P. falciparum, but the latter are slightly more specific because the HRP2 antigen may persist in blood for days or weeks after effective treatment. HRP2-detecting RDTs are not suitable for detecting treatment failure. RDTs are slightly less sensitive for detecting P. malariae and P. ovale. The WHO Malaria RDT Product Testing programme provides comparative data on the performance of RDT products to guide procurement. Since 2008, 210 products have been evaluated in five rounds of product testing (120)(123).

For the diagnosis of severe malaria, microscopy is preferred, as it provides a diagnosis of malaria and assessment of other important parameters of prognostic relevance in severely ill patients (such as parasite count and stage of parasite development and intra-leukocyte pigment). In severe malaria, an RDT can be used to confirm malaria rapidly so that parenteral antimalarial treatment can be started immediately. Where possible, however, blood smears should be examined by microscopy, with frequent monitoring of parasitaemia (e.g. every 12 h) during the first 2–3 days of treatment in order to monitor the response.

Choosing ACT

In the absence of resistance, all the recommended ACTs have been shown to result in parasitological cure rates of > 95%. Although there are minor differences in the oral absorption, bioavailability and tolerability of the different artemisinin derivatives, there is no evidence that these differences are clinically significant in currently available formulations. It is the properties of the partner medicine and the level of resistance to it that determine the efficacy of a formulation.

Policy-makers should also consider:

  • local data on the therapeutic efficacy of the ACT,
  • local data on drug resistance,
  • the adverse effect profiles of ACT partner drugs,
  • the availability of appropriate formulations to ensure adherence,
  • cost.

In parts of South-East Asia, artemisinin resistance is compromising the efficacy of ACTs and placing greater selection pressure on resistance to the partner medicines. Elsewhere, there is no convincing evidence for reduced susceptibility to the artemisinins; therefore, the performance of the partner drugs is the determining factor in the choice of ACT, and the following principles apply:

  • Resistance to mefloquine has been found in parts of mainland South-East Asia where this drug has been used intensively. Nevertheless, the combination with artesunate is very effective, unless there is also resistance to artemisinin. Resistance to both components has compromised the efficacy of artesunate + mefloquine in western Cambodia, eastern Myanmar and eastern Thailand.
  • Lumefantrine shares some cross-resistance with mefloquine, but this has not compromised its efficacy in any of the areas in which artemether + lumefantrine has been used outside South-East Asia.
  • Until recently, there was no evidence of resistance to piperaquine anywhere, but there is now reduced susceptibility in western Cambodia. Elsewhere, the dihydroartemisinin + piperaquine combination is highly effective.
  • Resistance to SP limits its use in combination with artesunate to the few areas in which susceptibility is retained.
  • Amodiaquine remains effective in combination with artesunate in parts of Africa and the Americas, although elsewhere resistance to this drug was prevalent before its introduction in an ACT.

Considerations in use of artemisinin-based combination therapy

Oral artemisinin and its derivatives (e.g. artesunate, artemether, dihydroartemisinin) should not be used alone. In order to simplify use, improve adherence and minimize the availability of oral artemisinin monotherapy, fixed-dose combination ACTs are strongly preferred to co-blistered or co-dispensed loose tablets and should be used when they are readily available. Fixed-dose combinations of all recommended ACT are now available, except artesunate + SP. Fixed-dose artesunate + amodiaquine performs better than loose tablets, presumably by ensuring adequate dosing. Unfortunately, paediatric formulations are not yet available for all ACTs.

The choice of ACT in a country or region should be based on optimal efficacy and adherence, which can be achieved by:

  • minimizing the number of formulations available for each recommended treatment regimen
  • using, where available, solid formulations instead of liquid formulations, even for young patients.

Although there are some minor differences in the oral absorption and bioavailability of different artemisinin derivatives, there is no evidence that such differences in currently available formulations are clinically significant. It is the pharmacokinetic properties of the partner medicine and the level of resistance to it that largely determine the efficacy and choice of combinations. Outside South-East Asia, there is no convincing evidence yet for reduced susceptibility to the artemisinins; therefore, the performance of the partner drug is the main determinant in the choice of ACT, according to the following principles:

  • Drugs used in IPTp, SMC or chemoprophylaxis should not be used as first-line treatment in the same country or region.
  • Resistance to SP limits use of artesunate + SP to areas in which susceptibility is retained. Thus, in the majority of malaria-endemic countries, first-line ACTs remain highly effective, although resistance patterns change over time and should be closely monitored.

Choosing among formulations

Use of fixed-dose combination formulations will ensure strict adherence to the central principle of combination therapy. Monotherapies should not be used, except as parenteral therapy for severe malaria or SP chemoprevention, and steps should be taken to reduce and remove their market availability. Fixed-dose combination formulations are now available for all recommended ACTs except artesunate + SP.

Paediatric formulations should allow accurate dosing without having to break tablets and should promote adherence by their acceptability to children. Paediatric formulations are currently available for artemether + lumefantrine, dihydroartemisinin + piperaquine and artesunate + mefloquine.

Other operational issues in managing effective treatment

Individual patients derive the maximum benefit from an ACT if they can access it within 24–48 h of the onset of malaria symptoms. The impact in reducing transmission at a population level depends on high coverage rates and the transmission intensity. Thus, to optimize the benefits of deploying ACTs, they should be available in the public health delivery system, the private sector and the community, with no financial or physical barrier to access. A strategy for ensuring full access (including community management of malaria in the context of integrated case management) must be based on analyses of national and local health systems and may require legislative changes and regulatory approval, with additional local adjustment as indicated by programme monitoring and operational research. To optimize the benefits of effective treatment, wide dissemination of national treatment guidelines, clear recommendations, appropriate information, education and communication materials, monitoring of the deployment process, access and coverage, and provision of adequately packaged antimalarial drugs are needed.

Community case management of malaria

Community case management is recommended by WHO to improve access to prompt, effective treatment of malaria episodes by trained community members living as close as possible to the patients. Use of ACTs in this context is feasible, acceptable and effective (173). Pre-referral treatment for severe malaria with rectal artesunate and use of RDTs are also recommended in this context. Community case management should be integrated into community management of childhood illnesses, which ensures coverage of priority childhood illnesses outside of health facilities.

Health education From the hospital to the community, education is vital to optimizing antimalarial treatment. Clear guidelines in the language understood by local users, posters, wall charts, educational videos and other teaching materials, public awareness campaigns, education and provision of information materials to shopkeepers and other dispensers can improve the understanding of malaria. They will increase the likelihood of better prescribing and adherence, appropriate referral and reduce unnecessary use of antimalarial medicines.

Adherence to treatment

Patient adherence is a major determinant of the response to antimalarial drugs, as most treatments are taken at home without medical supervision. Studies on adherence suggest that 3-day regimens of medicines such as ACTs are completed reasonably well, provided that patients or caregivers are given an adequate explanation at the time of prescribing or dispensing. Prescribers, shopkeepers and vendors should therefore give clear, comprehensible explanations of how to use the medicines. Co-formulation probably contributes importantly to adherence. User- friendly packaging (e.g. blister packs) also encourages completion of a treatment course and correct dosing.

Good practice statement

The choice of ACTs in a country or region should be based on optimal efficacy, safety and adherence.

Practical Info

Pharmacovigilance is the practice of monitoring the effects of medical drugs after they have been licensed for use, especially to identify and evaluate previously unreported adverse reactions. A practical handbook on the pharmacovigilance of antimalarial medicines (168) provides a step-by-step approach for antimalarial pharmacovigilance. Designed for health officials, planners, and other health workers, it focuses on active and passive pharmacovigilance, reporting, event monitoring and other key factors.

National adaptation and implementation (2015)

Good practice statement

Drugs used in IPTp, SMC and IPTi should not be used as a component of first- line treatments in the same country or region.

National adaptation and implementation (2015)

Good practice statement

When possible, use:

  • fixed-dose combinations rather than co-blistered or loose, single-agent formulations; and
  • for young children and infants, paediatric formulations, with a preference for solid formulations (e.g. dispersible tablets) rather than liquid formulations.

© World Health Organization 2022.

Sales, rights and licensing. To purchase WHO publications, see http://apps.who.int/bookorders. To submit requests for commercial use and queries on rights and licensing, see http://www.who.int/about/licensing.

Third-party materials. If you wish to reuse material from this work that is attributed to a third party, such as tables, figures or images, it is your responsibility to determine whether permission is needed for that reuse and to obtain permission from the copyright holder. The risk of claims resulting from infringement of any third-party-owned component in the work rests solely with the user.

Some rights reserved. This work is available under the Creative Commons AttributionNonCommercial-ShareAlike 3.0 IGO license (CC BY-NC-SA 3.0 IGO; https://creativecommons.org/licenses/by-nc-sa/3.0/igo).

Under the terms of this license, you may copy, redistribute and adapt the work for noncommercial purposes, provided the work is appropriately cited, as indicated below. In any use of this work, there should be no suggestion that WHO endorses any specific organization, products or services. The use of the WHO logo is not permitted. If you adapt the work, then you must license your work under the same or equivalent Creative Commons license. If you create a translation of this work, you should add the following disclaimer along with the suggested citation: “This translation was not created by the World Health Organization (WHO). WHO is not responsible for the content or accuracy of this translation. The original English edition shall be the binding and authentic edition”.

Any mediation relating to disputes arising under the license shall be conducted in accordance with the mediation rules of the World Intellectual Property Organization.

Bookshelf ID: NBK579027