Malaria

Published on June 2016 | Categories: Documents | Downloads: 34 | Comments: 0 | Views: 350
of 12
Download PDF   Embed   Report

Comments

Content

Seminar

Malaria
Brian M Greenwood, Kalifa Bojang, Christopher J M Whitty, Geoffrey A T Targett
Lancet 2005; 365: 1487–98 See Editorial page 1439

Malaria is the most important parasitic infection in people, accounting for more than 1 million deaths a year. Malaria has become a priority for the international health community and is now the focus of several new initiatives. Prevention and treatment of malaria could be greatly improved with existing methods if increased financial and labour resources were available. However, new approaches for prevention and treatment are needed. Several new drugs are under development, which are likely to be used in combinations to slow the spread of resistance, but the high cost of treatments would make sustainability difficult. Insecticide-treated bed-nets provide a simple but effective means of preventing malaria, especially with the development of longlasting nets in which insecticide is incorporated into the net fibres. One malaria vaccine, RTS,S/AS02, has shown promise in endemic areas and will shortly enter further trials. Other vaccines are being studied in clinical trials, but it will probably be at least 10 years before a malaria vaccine is ready for widespread use. Malaria is caused by protozoan parasites belonging to the genus Plasmodium. Four species account for almost all human infections (P falciparum, P vivax, P malariae, and P ovale). P falciparum causes the majority of infections in Africa and is responsible for most severe disease and mortality. P vivax and P ovale form resting stages in the liver (hypnozoites) that, once reactivated, can cause a clinical relapse many months after the initial event. Malaria can be transmitted by several species of female anopheline mosquitoes that differ in behaviour. This contributes to the varying epidemiological patterns of the disease seen worldwide. Because P vivax can develop in mosquitoes at a lower temperature than can P falciparum, its geographical range is wider. Figure 1 summarises the malaria life cycle. We present updated information about the challenges and opportunities for improved treatment and control. After World War II, strenuous efforts were made to eradicate malaria.1 Although these efforts were successful over large geographical areas, they did not succeed in tropical Africa or in many parts of Asia. The subsequent primary health-care initiative, which placed the main responsibility for malaria control on peripheral health-care workers, was equally unsuccessful. In the past few years, malaria has once again attracted more attention with the establishment of several new initiatives such as the Roll Back Malaria Partnership,2 partly because of increasing recognition that the malaria situation in sub-Saharan Africa has deteriorated during the past decade, for several reasons (panel 1).3 The importance of climatic warming is debated.4,5 War and civil unrest has led to an upsurge of malaria in many parts of Africa where health services have broken down.6 Malaria and HIV interact in several ways; malaria could adversely affect HIV infection by increasing viral load,7 whereas HIV increases malaria fevers8,9 and interacts adversely with malaria during pregnancy.10 However, the main cause of the worsened malaria situation recorded in recent years has been the spread of drug-resistant parasites, which has led to rising malaria-associated mortality, especially in east Africa, even though overall child mortality has fallen (figure 2).11,12
www.thelancet.com Vol 365 April 23, 2005

See Personal Account page 1499 Gates Malaria Partnership, Department of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1B 3DP, UK (Prof B M Greenwood FRS, K Bojang MD, C J M Whitty FRCP, Prof G A T Targett DSc); and Medical Research Council Laboratories, Banjul, The Gambia (K Bojang) Correspondence to: Prof Brian Greenwood [email protected]

The burden of malaria
To estimate accurately how many people die from malaria per year is important now that international organisations are setting specific targets for control programmes, but this measurement is difficult to achieve.13 Most deaths from malaria occur at home, so that information for these events can only come from use of the post-mortem questionnaire technique, an imprecise method. Attempts have been made to improve the reliability of this approach that take into account the sensitivity and specificity of the technique12 and combine epidemiological, geographic, and demographic data.14 Most estimates suggest that malaria directly causes about 1 million deaths per year or 3000 deaths a day, and that most of these deaths occur in African children (figure 3).14 Results of effective intervention studies suggest that the true number could be even higher because of indirect effects of the disease on nutrition and other infections. Accurate determination of the extent of morbidity caused by malaria is also difficult. Strenuous efforts are being made to measure the morbidity due to malaria by collation of all published reports and unpublished records by groups such as Malaria Risk Across Africa (MARA) and the Child Health Epidemiology Reference Group (CHERG).14–16 The consensus is that about 0·5 billion clinical attacks of malaria take place every year, including 2–3 million severe attacks (figure 3). In
Search strategy and selection criteria In addition to the review of key papers, we undertook searches of electronic databases. For PubMed, the search items were “malaria” restricted to the past 5 years, “malaria and trial”, “malaria and pathophysiology”, “malaria and diagnosis”, “malaria and vectors”, and “malaria and vaccines” without a time restriction. The Cochrane database of systematic reviews was searched by use of the term “malaria”. Articles were selected on the basis of their effect on malaria treatment or control. When more than one paper illustrated a specific point, the most representative paper was chosen.

1487

Seminar

Sporozoites

In mosquito gut Gamete zygote ookinete oocyst

Liver-stage parasites

Merozoites Gametocytes Ring

malaria does not occur,24 and that the disease costs Africa about US$12 billion every year. The burden of malaria falls disproportionately on poor and vulnerable individuals25 and its social consequences are poorly understood.26 Geographical information systems are being used to estimate the prevalence of malaria in areas that have no survey data by matching geographic features (obtained by satellite or ground-based systems) with those in areas where the epidemiological pattern of malaria is known.14,27 These information systems are also useful for the prediction of epidemics. Study of changes in global climatic patterns, such as El Niño activity, can provide medium-term warning of increased risk.28 At the local level, new data-collection systems are being developed to allow early detection of local outbreaks, thus providing an opportunity for the rapid implementation of control measures.29,30

The parasite
Malaria research has been transformed by the use of molecular genetic techniques.31 An example of the value of this investigative approach in the epidemiology of malaria has been the demonstration that a cluster of cases in Kapit (Sarawak, Malaysia) were caused by infection with the monkey parasite P knowlesi, and not with the morphologically similar P malariae as had been previously thought (figure 4).32
Panel 1: Reasons for deterioration of malaria control
G

Rupturing schizont Schizont

Trophozoite

Figure 1: Life cycle of the malaria parasite Adapted from Good MF. Vaccine-induced immunity to malaria parasites and the need for novel strategies. Trends Parasitol 2005; 21: 29–34.

malaria-endemic areas, malaria infection in pregnancy is believed to account for up to a quarter of all cases of severe maternal anaemia and for 10–20% of low birthweight babies.17,18 In addition to its direct effect in infants, the disease could account for an additional 5–10% of neonatal and infant deaths based on its effect on birthweight.19 Although most deaths from malaria arise in Africa, which justifies the focus of control efforts in Africa, evidence suggests that the number of clinical episodes of P falciparum malaria is higher than that widely quoted, and that morbidity due to malaria in Asia has been greatly underestimated,20 including that due to P vivax infections. The effect of malaria extends far beyond these direct measures of mortality and morbidity. Malaria can reduce attendance at school and productivity at work and evidence suggests that the disease can impair intellectual development.21 Cerebral malaria can result in persisting developmental abnormalities.22 The economic effect of malaria is immense;23 estimations during the past 35 years indicate that the yearly gross national product (GNP) has risen 2% less in countries where malaria remains highly endemic than in countries with an otherwise similar background where
1488

G

G

G

G

G

G

Climate instability: droughts and floods can increase malaria transmission in different epidemiological circumstances Global warming: global warming can increase transmission in some highland areas, but is unlikely to lead to a wide geographical spread of malaria Civil disturbances: civil unrest results in the collapse of malaria treatment programmes and crowding of refugees, some of whom might come from non-endemic areas, which enhances malaria transmission and can lead to epidemics Travel: increasing travel within endemic areas as well as by travellers from non-endemic to endemic areas puts many non-immune individuals at risk HIV: HIV increases susceptibility to malaria, raises the burden on the health services, and reduces the number of clinical staff available to treat malaria Drug resistance: drug resistance is probably the major cause of the deteriorating malaria situation in Africa Insecticide resistance: resistance to pyrethroids (used to treat bed-nets) has emerged in Anopheles gambiae in west Africa and in An funestus in southern Africa. Resistance in An gambiae is not yet severe enough to stop treated bednets from being effective, but resistance in An funestus is more substantial and had needed a change to DDT for household spraying in southern Africa

www.thelancet.com Vol 365 April 23, 2005

Seminar

Malaria mortality (1000 deaths per year)

14 12 10 8 6 4 2 0 60 50 40 30 20 10 0

1982–89 1990–98

Children

Pregnant women

+/–1

Deaths

? +/–0·5

Deaths Low birthweight Severe anaemia Infections

+/–5

Severe attacks +/–0·5

+/–250 100

Clinical attacks

+/–10 20

All-cause mortality (1000 deaths per year)

Figure 3: Number of malaria episodes and complications occurring every year in children younger than 5 years in sub-Saharan Africa and in African pregnant women Numbers are in millions. Severe attacks in children include about 1 million cases of cerebral malaria and 4 million cases of severe anaemia. Of children with clinical attacks, several thousand have neurological damage and up to 250 000 will have developmental problems. In pregnant women, low birthweight associated with anaemia is thought to contribute to 100 000 infant deaths every year.

West

East and south

Figure 2: Malaria mortality in Africa Adapted from reference 12, with permission from Elsevier.

Several studies have focused on genetic markers that characterise parasite virulence. The best studied of these is the P falciparum erythrocyte–membrane-protein-1 family that are responsible for antigenic variation and for cytoadherence of parasitised erythrocytes to endothelial cells and to cells of the placental syncytiotrophoblast. Parasites that cause severe malaria in nonimmune patients tend to express a small subset of these proteins that differ from those expressed by parasites that cause uncomplicated infections.33 There is also increasing evidence for the virulence of another subset of these variant proteins that mediate sequestration of P falciparum in the placenta.34 More virulence proteins are probably yet to be identified.35 Valuable information on the selection, transmission, and clearance of drug-resistant parasites has been obtained by the identification of molecular markers of drug resistance.36,37 Resistance to the antifolate drug combination sulfadoxine-pyrimethamine is linked to three or four mutations in the dihydrofolate reductase (dhfr) gene and to one or two mutations in the dihydropteroate synthase (dhps) gene. Surprisingly, these mutations have arisen infrequently and their current widespread distribution is due to gene flow.38 Resistance of P falciparum to chloroquine is largely due to mutations affecting the pfcrt and pfmdr1 genes that have also arisen only infrequently but are now widely distributed.39 Both chloroquine and sulfadoxinepyrimethamine have long half-lives; therefore, drugresistant parasites are at a selective advantage in communities where these drugs are widely used.40,41 However, in the absence of drug use, resistant parasites
www.thelancet.com Vol 365 April 23, 2005

seem not to be as well transmitted as wild-type parasites. Complete discontinuation of the use of these drugs (although not easy to achieve) might allow them to become useful again, especially if used in a combination.42 Vaccination with polymorphic proteins could lead to the selection of parasite populations that evade the immune response induced by the vaccine; this event can now be monitored by use of molecular techniques.43 Selection of parasite subpopulations was not reported in a trial44 of the pre-erythrocytic vaccine RTS,S/AS02 (discussed in detail later), but was recorded in a small trial45 of the blood-stage vaccine MSP (merozoite surface protein)-2 in Papua New Guinea.

Pathogenesis and clinical features of malaria
Although the clinical features of malaria have been welldescribed, severe malaria (predominantly due to P falciparum) is now known to be much more complicated

Figure 4: Blood film from a patient with malaria in Kapit, Sarawak Trophozoites and schizonts of P knowlesi, morphologically similar to those of P malariae, are shown.32

Courtesy of Balbir Singh

1489

Seminar

than originally thought. The clinical pattern of severe malaria differs between non-immune adults and semiimmune African children in several ways, with organ failure being more common in adults than children. The two most frequent presentations of severe malaria in African children are severe anaemia and cerebral malaria, but respiratory distress is the most dangerous, especially in combination with other syndromes.46 Cerebral malaria is the best-known form of severe malaria. Carefully undertaken post-mortem and electrophysiological studies in children have shown that this disease is a heterogeneous syndrome in which sequestration probably has a major role in some cases but little in others.47,48 Metabolic derangement including hypoglycaemia and subclinical convulsions are important in many cases. Sequestration is probably more consistently the cause in adults than in children.49 Severe malarial anaemia (figure 5) also consists of a group of conditions with different causes, including direct destruction of parasitised red blood cells, indirect destruction of non-parasitised red blood cells by immune mechanisms, and bone-marrow suppression associated with imbalances in cytokine concentrations.50 Pro-inflammatory cytokines such as tumour necrosis factor (TNF), nitric oxide, and various metabolic products, are associated with a poor prognosis in severe malaria, but whether this association is always causal is unclear.51,52 Acidosis is a good marker of disease severity. Initially, this event was thought to be due mainly to lactic acidosis but (in adults at least) lactic acidaemia now seems to be only one of several metabolic causes.53 In children, increasing evidence has shown that tissue hypoperfusion has a central role in disease severity; the relative importance of hypovolaemia and anaemia are uncertain.54,55 Research in this area is important, since fluid and (to a lesser extent) blood replacement are the two adjunctive therapies that are available even in poorly resourced hospitals. An extensive search for other adjunctive therapies based on current understanding of the pathophysiology of severe malaria has not yet yielded any product of established benefit, but further study is ongoing.

mortality due to bacterial disease is underestimated in febrile children.59,60 In adults living in areas where HIV prevalence is high, much of the severe illness treated as malaria is probably HIV-related, although this assumption is not yet proven.61 Algorithms that attempt to differentiate malaria from other causes of febrile illness in young children using clinical features alone have not proved sensitive or specific enough to guide treatment62 and, where malaria is endemic but parasitological diagnosis cannot be undertaken, children with fever should be given antimalaria treatment according to Integrated Management of Childhood Illness (IMCI) guidelines, even if another diagnosis such as pneumonia is regarded as likely. Accurate microscopy can be helpful in diagnosis, but maintenance of the microscopes and the quality of microscopy in peripheral clinics is difficult.63 Alternatively, several rapid diagnostic tests based on antigen-capture techniques have been developed that have high sensitivity and specificity for falciparum malaria and that could contribute greatly to improving malaria diagnosis in some situations.64,65 However, these tests have limitations. In highly malaria-endemic areas, many healthy individuals have parasitaemia; thus, although a negative test rules out malaria, a positive test does not prove that malaria is the cause of illness. Another limitation of rapid diagnostic tests is their cost. The introduction of artemisinin-based combination therapy (ACT) probably will make rapid diagnostic tests more cost-effective, but only if their use leads to a substantial reduction in overprescription of antimalarial drugs. Until recently, first-line treatment for falciparum and vivax malaria was the same (ie, chloroquine) in poorly resourced parts of Asia, such as Afghanistan, where both infections are found and where diagnostic services are scarce. Because P falciparum but not P vivax has become highly resistant to chloroquine in many of these areas, treatment for both infections is diverging and species diagnosis has now become increasingly important. Unfortunately, most rapid diagnostic tests do not detect vivax infections, and those that do are expensive.

Diagnosis of malaria
In tropical Africa, many patients treated for mild or severe malaria do not actually have the disease,56,57 especially in adults diagnosed as having cerebral malaria.58 Conversely many cases of malaria are not diagnosed. Overdiagnosis and consequent overprescription of antimalarial drugs for the treatment of uncomplicated malaria was not a major problem when chloroquine was used as first-line treatment, since the drug was safe and cheap. However, over-diagnosis has become a major issue now that drug combinations, which are expensive and in relatively short supply, are the preferred form of first-line treatment. The issue of most concern for patients with severe febrile illness is that treatable alternative diagnoses are being missed. Good evidence now indicates that in Kenya,
1490

Figure 5: Gambian child with severe malaria anaemia

www.thelancet.com Vol 365 April 23, 2005

Courtesy of Martin Weber

Seminar

Non-artemisinin combinations Quinine and sulfadoxine-pyrimethamine Quinine and doxycycline Sulfadoxine-pyrimethamine and chloroquine Sulfadoxine-pyrimethamine and amodiaquine

Used effectively in Europe and parts of Asia. Long treatment course, cost and side-effects make combination inappropriate for the African market Similar to quinine and sulfadoxine-pyrimethamine, mainly used where sulfadoxine-pyrimethamine resistance is a problem (eg, historically in Thailand) Current policy used in some African countries, but is ineffective where resistance to both drugs is high Substantially more effective than sulfadoxine-pyrimethamine and chloroquine in areas where amodiaquine resistance is low

Artemisinin-based combination treatments (ACTs) Artemether-lumefantrine Currently the only internationally licenced coformulated ACT. Available in Asia and Africa Artesunate and amodiaquine Currently copackaged. Adopted as policy by some African counties. Effective where amodiaquine resistance is low Dihydroartemisinin-piperaquine Coformulated drug that has been used widely in Asia and is presently being assessed in a new formulation for licensing Artesunate and mefloquine Mainstay of antimalarial drug policy in much of southeast Asia. Regarded as too expensive for the African market Artesunate and sulfadoxine-pyrimethamine Treatment used in some Asian countries (eg, Afghanistan). Ineffective where sulfadoxine-pyrimethamine has failed Dihydroartemisinin-napthoquine-trimethoprim New formulation used in China and Vietnam. Early reports are encouraging

Table: Examples of currently used combinations of antimalarial drugs

Treatment of malaria
Although chloroquine is still the first-line treatment for uncomplicated falciparum malaria in some countries, it now fails almost everywhere. Resistance to sulfadoxinepyrimethamine has occurred in most countries where the drug has been introduced to replace chloroquine,66 although not in all.67 Some form of combination treatment is now clearly needed for the first-line treatment of falciparum malaria in Africa and Asia,68 and in some countries a switch to combination treatment is well overdue. Combination therapy with drugs with different modes of action is now the preferred approach to malaria treatment to inhibit the emergence and spread of parasites resistant to one component of the combination. What combination of drugs should be used and how can such treatment be afforded? Combinations of drugs with similar half-lives are desirable so that parasites are not exposed to one component of the combination alone for long periods. The most widely promoted combinations are ACTs (table). A coformulated ACT, artemether-lumefantrine, is already available with good efficacy data from Asia69 and promising effectiveness data from east Africa,70,71 although few published data support its use in some of the areas where its use is being considered.72 A combination of artesunate and amodiaquine works well in areas where resistance to amodiaquine is modest.73 In Africa, the addition of artemisinin to a drug that has failed locally only leads to a failed combination.71,74 However, in southeast Asia, mefloquine resistance might have been reversed by combination with artemisinin.75 Not all effective combinations have to contain artemisinin. The combination of amodiaquine and sulfadoxine-pyrimethamine is still efficacious in some areas of Africa where only moderate resistance to these drugs exists,76 but will remain so only if the component drugs are not used widely for monotherapy. Although the current choice of ACTs is restricted, the future is encouraging, with several new drugs at various stages of development.77 Two have considerable promise.
www.thelancet.com Vol 365 April 23, 2005

First, the fixed-dose combination dihydroartemisininpiperaquine is already being used in much of southeast Asia and has proved very efficacious.78 The second treatment, which is being designed for the African market where drugs must be cheap as well as effective, is a combination of artesunate and chlorproguanildapsone. The combination is cheap, well-tolerated, and works well even in areas where sulfadoxinepyrimethamine has failed.79,80 Questions about the safety of artemisinin in pregnancy81 and of chlorproguanildapsone in those with glucose-6-dehydrogenase deficiency are being addressed. Other new drugs, including those that are modifications of existing antimalarial classes and others with a novel mode of action, are entering clinical trials and the prospects for effective new treatments are much more encouraging than they were a few years ago.77 The major obstacle to large-scale use of ACTs is their cost; they are up to ten times more expensive than current monotherapy, which is unrealistic in many settings unless subsidies are introduced.82 A decision to encourage most African countries to change to ACTs simultaneously, irrespective of their immediate need, has contributed to a short-term quadrupling in the cost

See Articles pages 1467 and 1474

Figure 6: Artemisinin suppositories for early management of severe malaria in a village in northern Ghana

1491

Courtesy of Brian Greenwood

Seminar

of artemisinins and a global shortage of raw materials. High costs of ACTs have encouraged the production of counterfeits.83 Synthetic versions of artemisinins (that are being developed) could reduce costs,84 but in the medium-term at least, improvement of the supply of Artemisia and substantial subsidies for ACTs are the only realistic options85 if ACTs are to be used widely in the many parts of Asia and Africa where the treatment is needed most. Many deaths from malaria occur in the community before any contact is made with the formal health sector. Several developments have the potential to improve this situation. These advances include home-based management that provides effective treatment as close to home as possible,86 training of shopkeepers who sell drugs,87 packaging of drugs,88 and development of artemisinin suppositories, which can be given to the community for initial treatment of severe malaria before transfer to a treatment centre (figure 6).89 Achievement of a good balance between the provision of presumptive treatment close to home (to ensure rapid treatment) and the reduction in overprescription of increasingly costly antimalarial drugs is a major challenge.

Panel 2: Key questions to be answered before IPTi can be recommended for routine implementation
G G

G

G

G

G

Is IPTi safe? Will the administration of drugs at the time of vaccination impair the immune response to routine vaccines? Does IPTi require a longlasting drug and, if this is the case, which drug could be used to replace sulfphadoxinepyrimethamine in areas where this drug is losing efficacy? What effect will IPTi have on the development of natural immunity to malaria? Will implementation of IPTi encourage or discourage parents from bringing their children for vaccination? Can IPTi be introduced into routine EPI (Expanded Programme on Immunization [WHO]) clinics without causing disruption?

Antimalarial drugs and the prevention of malaria
See Research Letters page 1481

Several million non-immune people travel to malariaendemic areas every year for business or pleasure, and they need to be protected. The decision on the most appropriate drug for particular travellers to take for chemoprophylaxis needs a detailed knowledge of their medical history and of the places they will visit. Guidelines on chemoprophylaxis for travellers are provided by several national and international organisations.90,91 Early studies in endemic areas showed that chemoprophylaxis during pregnancy reduced the incidence of severe maternal anaemia and improved birthweight. In children, chemoprophylaxis lowered mortality and morbidity from malaria substantially, but it is difficult to sustain over long periods, could encourage drug resistance, and could impede the development of natural immunity.92 Thus, the discovery that sulfadoxinepyrimethamine given on two or three occasions during pregnancy was more effective at preventing infection of the placenta than chemoprophylaxis with chloroquine was a breakthrough.93 Subsequent studies have shown that this approach, now known as intermittent preventive therapy in pregnancy (IPTp), protects against maternal anaemia94 and low birthweight,95 especially in primigravidae and secundigravidae, and its use in areas of medium or high transmission is recommended by WHO. Unfortunately, the efficacy of IPTp is reduced in HIV-positive women.96 Whether IPTp with sulfadoxinepyrimethamine is effective through prevention of infection or intermittent clearance of parasites from the placenta is not known. This issue is important since

increasing resistance to sulfadoxine-pyrimethamine makes the search more urgent for a safe and effective alternative therapy. IPT has also been used to protect children. Two studies undertaken in Tanzania97,98 showed that use of sulfadoxine-pyrimethamine or amodiaquine at specific times during the first year of life (IPTi) lowered the incidence of malaria and severe anaemia without any rebound in clinical malaria the following year.99 These encouraging results have led to the creation of an IPTi consortium addressing several important issues related to the approach (panel 2).100 In areas where malaria is highly seasonal and affects older children, a different approach to IPT is needed. Preliminary results are encouraging from studies in Senegal and Mali, in which antimalarial drugs were given to children (younger than 5 years) at set times during the short transmission season.101,102 IPT is also being investigated as a means of protecting other groups at risk, such as children with anaemia.103 Vitamin and mineral supplements have been used to reduce the incidence of clinical attacks of malaria with success for vitamin A,104 but with conflicting results for zinc.105,106

Vector control
The genome of Anopheles gambiae has now been sequenced,107 providing opportunities for new, targeted measures for control. For example, use of olfactory cues by female mosquitoes to find their human hosts and identification of odorant-binding proteins in the antennae of mosquitoes that respond to components of human sweat could lead to development of new types of insect traps or repellents.108 A 20-year programme aims to develop and release mosquitoes that are fully refractory to Plasmodium. Although refractory strains have been produced in laboratories, the challenge is to find ways to drive resistance mechanisms into the wild mosquito population, without the genetically altered
www.thelancet.com Vol 365 April 23, 2005

1492

Seminar

mosquitoes being at a selective disadvantage and being quickly eliminated.109 Alternative genetic modifications that are lethal to female mosquitoes could be more effective.110 Mosquito control has been at the centre of past efforts to eradicate malaria, mainly through the use of the insecticide DDT (dichloro-diphenyl-trichloroethane) for indoor residual house-spraying. Notable successes were achieved, but the programme was not sustainable. Despite attempts to ban DDT completely, the compound continues to be used for vector control although arguments about issues such as the effect on preterm births and the duration of lactation continue.111 In some southern African countries (mainly those with unstable patterns of malaria), indoor house-spraying with DDT, carbamates, or pyrethroids, used alone or with ACTs has substantially improved malaria control.112 Trials of insecticide-treated nets (ITNs) have consistently shown reductions in overall child mortality and in episodes of clinical malaria during a 1–2 year period.113 In a major study in Kenya,114 there were substantial health gains in both children and pregnant women, and protection extended from homes with ITNs to adjacent homes without nets.115 Encouragingly, use of ITNs for 6–7 years has not shown a shift in child mortality from younger to older children.116,117 Regular re-treatment of nets with insecticide has proved difficult to sustain on a large scale, especially if users are required to pay for it. This problem should be overcome by the development of longlasting insecticidal nets, in which insecticide is incorporated into the net fibres. Different prototypes are being produced, two of which have now been approved by WHO and are undergoing large-scale production.118 In emergency situations, insecticide-treated tarpaulins have proved very effective (figure 7), and materials that have long-acting insecticidal action are being developed for use in this way. Repellents could provide useful protection against malaria,119 especially in places where vector mosquitos bite early in the evening. In such situations they might add benefit to ITNs. Inevitably, resistance to pyrethroids is emerging,120 but progress is being made in the identification of alternative insecticides that could be used to treat nets and other materials.121 One approach to prevent insecticide resistance is the use of mixtures or mosaics of insecticides on nets. This method would be difficult to sustain if repeated re-treatment of nets was needed, but trials of this process would be worthwhile for longlasting insecticidal nets. Despite the proven benefits and cost-effectiveness of ITNs, achievement of widespread use has proved difficult. In most sub-Saharan countries, only a small percentage of individuals who should be protected by nets actually use them.122 Full cost recovery for nets and insecticides makes them unavailable to the most vulnerable groups. Part cost recovery by use of vouchers and often combined with social marketing123 solves the
www.thelancet.com Vol 365 April 23, 2005

Figure 7: Insecticide-treated tarpaulins used for malaria prevention in a refugee camp in Sierra Leone (Tobanda)

problem to some extent. Free provision of ITNs, sometimes linked to other initiatives such as vaccination or attendance to antenatal clinics, has strong advocates124 and is gaining support, but this approach needs a major and sustained commitment from international donors.125 Environmental management (including drainage of breeding sites), improvements to house design, use of larvivorous fish, and zooprophylaxis have proved effective in some specific epidemiological situations but must be based on detailed behavioural knowledge of the main local vectors.

Malaria vaccines
Vaccine development against both falciparum and vivax malaria is ongoing.126,127 The decision on which parasite antigens are to undergo clinical development is difficult. Criteria for development include evidence showing that the antigen serves a function critical to the parasite, is associated with naturally acquired immunity, or is protective in animal models.128 So far, most efforts have been directed at the development of pre-erythrocytic stage vaccines designed to prevent invasion of hepatocytes by sporozoites or to destroy infected hepatocytes (figure 8). RTS,S/AS02A is the most advanced pre-erythrocytic vaccine, and is a hybrid molecule in which the circumsporozoite protein of P falciparum is expressed with hepatitis-B surface antigen (HBsAg) in yeast.129 The vaccine is given with AS02, a complex three-component adjuvant. RTS,S/ AS02A has provided substantial, short-lived protection in volunteers, exposed experimentally to bites by infected mosquitoes,130 and substantial (71%) but only short-term protection in naturally exposed, semi-immune adults from The Gambia.131 Some protection was restored by one booster dose of vaccine given the next year. In a subsequent trial in Mozambican children,132 RTS,S/AS02A gave 30% protection against the first clinical episode of malaria and 58% protection against severe malaria. Comparison of different methods of immunisation with pre-erythrocytic vaccines has shown that schedules that
1493

Courtesy of Matthew Burns

Seminar

Pre-erythrocytic stages Sporozoites CSP TRAP Asexual blood stages Merozoites MSP-1, 2, 3, 4, 5 AMA-1 EBA-175 Schizonts Pf EMP-1 Trophozoites RESA GLURP Sexual stages Human Macrogametocyte Mosquito Macrogamete Pfs 230 Pfs 48/45 Microgametocyte Microgamete Oocyst Zygote /ookinete Pfs 25/Pfs 28 Pvs 25/Pvs 28 (ME) Exo-erythrocytic (liver) stage LSA-1,3 EXP-1

blood stage. Surprisingly a combination of very low doses of infection and drug treatment gave complete protection against challenge in volunteers.138 Transmission-blocking vaccines are designed to prevent mosquitoes that feed on vaccinated individuals from becoming infected, reducing transmission and providing indirect protection to the entire population.139 Transmission-blocking vaccines against P falciparum and P vivax are well in advance and phase I trials are taking place. Malarial vaccine research has progressed rapidly over the past few years, helped by the availability of more funds and by improved organisation mediated through organisations such as the Malaria Vaccine Initiative.140 However, it is likely to be at least a decade before an efficacious vaccine is available for widespread use in malaria-endemic countries.

Conclusions
There is no clear, single path to improve malaria control. Such an approach will probably come from a series of incremental steps involving better and more widespread use of the methods that have already been shown to be effective, as well as the step-wise introduction of new treatments and partly effective control measures shown to be of benefit. Establishment of an effective malaria control programme—eg, one based on ACTs for treatment of diagnosed cases or on ITNs and IPTp for prevention— costs about $2–5 per person per year, a month’s wages for many average-sized families. Thus, if wide coverage is to be obtained with these simple interventions, subsidies will be needed for poor and vulnerable individuals. For the first time, the international health community could be prepared to make this possible. The formation of the Global Fund Against AIDS, Tuberculosis and Malaria was an important first step in this direction and the group has committed nearly $1 billion to malaria control for the next 2 years.141 It is essential that the international community continues to support this initiative. Although much more can be done with existing measures, new methods for malaria control are needed. Development of new antimalarial drugs or malaria vaccines is not an attractive financial option for large pharmaceutical companies. Thus, the creation of publicprivate partnerships such as the Medicines for Malaria Venture77 and the Malaria Vaccine Initiative,140 in which major pharmaceutical companies can participate, is an important step forward. New drugs, vaccines, and vector control measures will need evaluation, both alone and, increasingly, in combination, which will need innovative research designs. Such studies, carried out to full international standards will have to be done in malariaendemic areas. Currently few centres in endemic areas have the infrastructure or staff to take this on. Increased investment in more centres and scientists from malariaendemic countries is urgent.
www.thelancet.com Vol 365 April 23, 2005

Figure 8: Malaria vaccines under development CSP=circumsporozoite protein. TRAP=thrombospondin-related adhesion protein. LSA=liver-stage antigen. EXP=exported antigen. ME=multiple-epitope string based on T-cell and B-cell epitopes of pre-erythrocytic stage antigens. MSP=merozoite surface protein. AMA=apical merozoite antigen. EBA=erythrocyte-binding antigen. RESA=ring-infected erythrocyte surface antigen. GLURP=glutamine-rich protein. Pf EMP=P falciparum erythrocytemembrane protein. Pfs 230 and Pfs 48/45=antigens located on the surface of P falciparum gametocytes. Pfs 25/Pfs 28 and Pvs 25/Pvs 28=antigens located on the surface of ookinetes of P falciparum and P vivax, respectively.

use different presentations of the same antigen for different doses (prime-boost immunisation) are especially effective in inducing T-cell responses.133,134 DNA, modified vaccinia and fowlpox vaccines encoding identical DNA sequences of epitopes from preerythrocytic antigens, and the entire sequence of the thrombospondin-related adhesion protein (TRAP) have undergone clinical trials. Several phase I studies have generated data showing the safety and strong immunogenicity of these vaccines.135 Results of a phase IIb trial in semi-immune adult men in The Gambia were disappointing,136 but additional trials based on this strategy, including a phase IIb trial in Kenyan children, are ongoing. The aim of blood-stage vaccines is to eliminate or reduce the number of blood-stage parasites. Efforts to develop blood-stage vaccines have focused mainly on antigens contributing to erythrocyte invasion (figure 8).137 A combination vaccine of three blood-stage antigens reduced parasite density in a strain-specific manner but had no pronounced effect on the overall number of clinical malaria episodes.45 Vaccines based on the bloodstage vaccines MSP-1 and AMA (apical merozoite antigen)-1 are currently undergoing clinical trials. Multicomponent vaccines will probably be needed to cope with the problem of antigenic polymorphism. An alternative is whole-organism vaccines—sporozoite or
1494

Seminar

Major efforts are being made to develop new procedures for malaria treatment and control. However, these approaches will only achieve their maximum potential if a functioning health-care system can deliver them.142 Improved treatment throughout the malariaendemic areas of Africa will need a major increase in staff numbers, provision of appropriate training, and provision of sufficient monetary and non-financial incentives to retain key staff in post.
Conflict of interest statement We declare that we have no conflict of interest. Acknowledgments All the authors are supported by the Gates Malaria Partnership, which receives financial support from the Bill and Melinda Gates Foundation. References 1 Litsios S. The tomorrow of malaria. Wellington: Pacific Press 1996. 2 The Roll Back Malaria Partnership. http://www.rbm.who.int (accessed Jan 23, 2005). 3 Greenwood B, Mutabingwa T. Malaria in 2002. Nature 2002; 415: 670–72. 4 Hay SI, Rogers DJ, Randolph SE, et al. Hot topic or hot air? Climate change and malaria resurgence in East African highlands. Trends Parasitol 2002; 18: 530–34. 5 Tanser FC, Sharp B, le Sueur D. Potential effect of climate change on malaria transmission in Africa. Lancet 2003; 362: 1792–98. 6 Bloland PB, Williams HA. Malaria control during mass population movements and natural disasters. National Academies Press, Washington, 2003. 7 Kublin JG, Patnaik P, Jere CS, et al. Effect of Plasmodium falciparum malaria on concentration of HIV-1-RNA in the blood of adults in rural Malawi: a prospective cohort study. Lancet 2005; 365: 233–40. 8 Whitworth J, Morgan D, Quigley M, et al. Effect of HIV-1 and increasing immunosuppression on malaria parasitaemia and clinical episodes in adults in rural Uganda: a cohort study. Lancet 2000; 356: 1051–56. 9 French N, Nakiyingi J, Lugada E, et al. Increasing rates of malarial fever with deteriorating immune status in HIV-1-infected Ugandan adults. AIDS 2001; 15: 899–906. 10 Mount AM, Mwapasa V, Elliott SR, et al. Impairment of humoral immunity to Plasmodium falciparum malaria in pregnancy by HIV infection. Lancet 2004; 363: 1860–67. 11 Snow RW, Trape J-F, Marsh K. The past, present and future of childhood malaria mortality in Africa. Trends Parasitol 2001; 17: 593–97. 12 Korenromp EL, Williams BG, Gouws E, Dye C, Snow RW. Measurement of trends in childhood malaria mortality in Africa: an assessment of progress toward targets based on verbal autopsy. Lancet Infect Dis 2003; 3: 349–58. 13 de Savigny D, Binka F. Monitoring future impact on malaria burden in sub-Saharan Africa. Am J Trop Med Hyg 2004; 71 (suppl 2): 224–31. 14 Snow RW, Guerra CA, Noor AM, Myint HY, Hay SI. The global distribution of clinical episodes of Plasmodium falciparum malaria. Nature 2005; 434: 214–17. 15 Mapping Malaria Risk in Africa. http://www.mara.org.za (accessed Jan 23, 2005). 16 London School of Hygiene and Tropical Medicine. Deriving mortality and morbidity estimates for common childhood diseases. http://www.lshtm.ac.uk/nphiru/research/cherg.html (accessed Jan 23, 2005). 17 Steketee RW, Nahlen BL, Parise ME, Menendez C. The burden of malaria in pregnancy in malaria-endemic areas. Am J Trop Med Hyg 2001; 64 (1–2 suppl): 28–35. 18 Guyatt HL, Snow RW. The epidemiology and burden of Plasmodium falciparum related anemia among pregnant women in sub-Saharan Africa. Am J Trop Med Hyg 2001; 64 (1–2 suppl): 36–44.

19

20

21

22

23

24 25

26

27 28 29

30

31 32

33

34

35

36

37

38

39 40

41

42

Guyatt HL, Snow RW. Malaria in pregnancy as an indirect cause of infant mortality in sub-Saharan Africa. Trans R Soc Trop Med Hyg 2001; 95: 569–76. Hay SI, Guerra CA, Tatem AJ, Noor AM, Snow RW. The global distribution and population at risk of malaria: past, present, and future. Lancet Infect Dis 2004; 4: 327–36. Fernando SD, Gunawardena DM, Bandara MR, et al. The impact of repeated malaria attacks on the school performance of children. Am J Trop Med Hyg 2003; 69: 582–88. Carter JA, Ross AJ, Neville BG, et al. Developmental impairments following severe falciparum malaria in children. Trop Med Int Health 2005; 10: 3–10. Chima RI, Goodman CA, Mills A. The economic impact of malaria in Africa: a critical review of the evidence. Health Policy 2003; 63: 17–36. Sachs J, Malaney P. The economic and social burden of malaria. Nature 2002; 415: 680–85. Barat LM, Palmer N, Basu S, Worrall E, Hanson K, Mills A. Do malaria control interventions reach the poor? A view through the equity lens. Am J Trop Med Hyg 2004; 71 (suppl 2): 174–78. Jones COH, Williams HA. The social burden of malaria: what are we measuring? Am J Trop Med Hyg 2004; 71 (suppl 2): 156–61. Rogers DJ, Randolph SE, Snow RW, Hay SI. Satellite imagery in the study and forecast of malaria. Nature 2002; 415: 710–15. Thomson MC, Connor SJ. The development of malaria early warning systems for Africa. Trends Parasitol 2001; 17: 438–45. Abeku TA, Hay SI, Ochola S, et al. Malaria epidemic early warning and detection in African highlands. Trends Parasitol 2004; 20: 400–05. Teklehaimanot HD, Schwartz J, Teklehaimanot A, Lipsitch M. Alert threshold algorithms and malaria epidemic detection. Emerg Infect Dis 2004; 10: 1220–26. Vernick KD, Waters A. Genomics and malaria control. N Engl J Med 2004; 351: 1901–04. Singh B, Kim Sung L, Matusop A, et al. A large focus of naturally acquired Plasmodium knowlesi infections in human beings. Lancet 2004; 363: 1017–24. Jensen ATR, Magistrado P, Sharp S, et al. Plasmodium falciparum associated with severe childhood malaria preferentially expresses PfEMPI encoded by group A var genes. J Exp Med 2004; 199: 1179–90. Staalsoe T, Shulman CE, Bulmer JN, Kawuondo K, Marsh K, Hviid L. Variant surface antigen-specific IgG and protection against clinical consequences of pregnancy-associated Plasmodium falciparum malaria. Lancet 2004; 363: 283–89. Marti M, Good RT, Rug M, Kneupfer E, Cowman AF. Targeting malaria virulence and remodeling proteins to the host erythrocyte. Science 2004; 306: 1930–33. Djimdé AA, Doumbo AK, Traore et al. Clearance of drug-resistant parasites as a model for protective immunity in Plasmodium falciparum malaria. Am J Trop Med Hyg 2003; 69: 559–63. Sutherland CJ, Alloueche A, Curtis J, et al. Gambian children successfully treated with chloroquine can harbor and transmit Plasmodium falciparum gametocytes carrying resistance genes. Am J Trop Med Hyg 2002; 67: 578–85. Roper C, Pierce R, Bredenkamp B, et al. Antifolate antimalarial resistance in southeast Africa: a population-based analysis. Lancet 2003; 361: 1174–81. Hastings IM. The origins of antimalarial drug resistance. Trends Parasitol 2004; 20: 512–18. Hallett RL, Sutherland CJ, Alexander N, et al. Combination therapy counteracts the enhanced transmission of drug-resistant malaria parasites to mosquitoes. Antimicrob Agents Chemother 2004; 48: 3940–43. Hastings IM, Watkins WM, White NJ. The evolution of drug resistant malaria: the role of drug elimination half-life. Philos Trans R Soc Lond B Biol Sci 2002; 357: 505–19. Mita T, Kaneko A, Lum JK, et al. Recovery of chloroquine sensitivity and low prevalence of the Plasmodium falciparum chloroquine resistance transporter gene mutation K76T following the discontinuance of chloroquine use in Malawi. Am J Trop Med Hyg 2003; 68: 413–15.

www.thelancet.com Vol 365 April 23, 2005

1495

Seminar

43

44

45

46

47

48

49

50 51

52

53

54 55 56

57

58

59

60

61

62

63 64

65

Felger I, Genton B, Smith T, Tanner M, Beck H-P. Molecular monitoring in malaria vaccine trials. Trends Parasitol 2003; 19: 60–63. Alloueche A, Milligan P, Conway DJ, et al. Protective efficacy of the RTS,S/ASO2 Plasmodium falciparum malaria vaccine is not strain specific. Am J Trop Med Hyg 2003; 68: 97–101. Genton B, Betuela I, Felger I, et al. A recombinant blood-stage vaccine reduces Plasmodium falciparum density and exerts selective pressure on parasite populations in a Phase 1–2b trial in Papua New Guinea. J Infect Dis 2002; 185: 820–27. Schellenberg D, Menendez C, Kahigwa E, et al. African children with malaria in an area of intense Plasmodium falciparum transmission: features on admission to the hospital and risk factors for death. Am J Trop Med Hyg 1999; 61: 431–38. Taylor TE, Fu WJ, Carr RA, et al. Differentiating the pathologies of cerebral malaria by postmortem parasite counts. Nat Med 2004; 10: 143–45. Crawley J, Smith S, Muthinji P, Marsh K, Kirkham F. Electroencephalographic and clinical features of cerebral malaria. Arch Dis Child 2001; 84: 247–53. Silamut K, Phu NH, Whitty C, et al. A quantitative analysis of the microvascular sequestration of malaria parasites in the human brain. Am J Pathol 1999; 155: 395–410. Ekvall H. Malaria and anemia. Curr Opin Hematol 2003; 10: 108–14. Boutlis CS, Weinberg JB, Baker J, et al. Nitric oxide production and nitric oxide synthase activity in malaria-exposed Papua New Guinean children and adults show longitudinal stability and no association with parasitemia. Infect Immun 2004; 72: 6932–38. Lyke KE, Burges R, Cissoko Y, et al. Serum levels of the proinflammatory cytokines interleukin-1 beta (IL-1beta), IL-6, IL-8, IL-10, tumor necrosis factor alpha, and IL-12(p70) in Malian children with severe Plasmodium falciparum malaria and matched uncomplicated malaria or healthy controls. Infect Immun 2004; 72: 5630–37. Dondorp AM, Chau TT, Phu NH, et al. Unidentified acids of strong prognostic significance in severe malaria. Crit Care Med 2004; 32: 1683–88. Maitland K, Newton CR. Acidosis of severe falciparum malaria: heading for a shock? Trends Parasitol 2005; 21: 11–16. Planche T, Onanga M, Schwenk A, et al. Assessment of volume depletion in children with malaria. PLoS Med 2004; 1: e18. Amexo M, Tolhurst R, Barnish G, Bates I. Malaria misdiagnosis: effects on the poor and vulnerable. Lancet 2004; 364: 1896–98. Reyburn H, Mbatia R, Drakeley C, et al. Overdiagnosis of malaria in patients with severe febrile illness in Tanzania: a prospective study. BMJ 2004; 329: 1212–15. Makani J, Matuja W, Liyombo E, Snow RW, Marsh K, Warrell DA. Admission diagnosis of cerebral malaria in adults in an endemic area of Tanzania: implications and clinical description. QJM 2003; 96: 355–62. Berkley JA, Lowe BS, Mwangi I, et al. Bacteremia among children admitted to a rural hospital in Kenya. N Engl J Med 2005; 352: 39–47. Evans JA, Adusei A, Timmann C, et al. High mortality of infant bacteraemia clinically indistinguishable from severe malaria. QJM 2004; 97: 591–97. Peters RP, Zijlstra EE, Schijffelen MJ, et al. A prospective study of bloodstream infections as cause of fever in Malawi: clinical predictors and implications for management. Trop Med Int Health 2004; 9: 928–34. Chandramohan D, Jaffar S, Greenwood BM. Use of clinical algorithms for diagnosing malaria. Trop Med Int Health 2002; 7: 45–52. Bates I, Bekoe V, Asamoa-Adu A. Improving the accuracy of malaria-related laboratory tests in Ghana. Malar J 2004; 3: 38. Craig MH, Bredenkamp BL, Williams CH et al. Field and laboratory comparative evaluation of ten rapid malaria diagnostic tests. Trans R Soc Trop Med Hyg 2002; 96: 258–65. Moody A. Rapid diagnostic tests for malaria parasites. Clin Microbiol Rev 2002; 15: 66–78.

66

67

68 69

70

71

72

73

74

75

76

77 78

79

80

81 82

83

84

85

East African Network for Monitoring Antimalarial Treatment (EANMAT). The efficacy of antimalarial monotherapies, sulphadoxine-pyrimethamine and amodiaquine in East Africa: implications for sub-regional policy. Trop Med Int Health 2003; 8: 860–67. Plowe CV, Kublin JG, Dzinjalamala FK, et al. Sustained clinical efficacy of sulfadoxine-pyrimethamine for uncomplicated falciparum malaria in Malawi after 10 years as first line treatment: five year prospective study. BMJ 2004; 328: 545–48. Kremsner PG, Krishna S. Antimalarial combinations. Lancet 2004; 364: 285–94. Mayxay M, Khanthavong M, Lindegardh N, et al. Randomized comparison of chloroquine plus sulfadoxine-pyrimethamine versus artesunate plus mefloquine versus artemether-lumefantrine in the treatment of uncomplicated falciparum malaria in the Lao People’s Democratic Republic. Clin Infect Dis 2004; 39: 1139–47. Piola P, Fogg C, Bajunirwe F, et al. Supervised versus unsupervised intake of six-dose artemether-lumefantrine for the treatment of acute, uncomplicated Plasmodium falciparum malaria in Mbarara, Uganda: a randomised trial. Lancet 2005; 365: 1467–73. Mutabingwa T, Anthony D, Heller A, et al. Amodiaquine alone, amodiaquine sulfadoxine-pyrimethamine, amodiaquine artesunate, and artemether-lumefantrine for outpatient treatment of malaria in Tanzanian children: a four-arm randomised effectiveness trial. Lancet 2005; 365: 1474–80. Omari AA, Preston C, Garner P. Artemether-lumefantrine for treating uncomplicated falciparum malaria. Cochrane Database Syst Rev 2003; 2: CD003125. Adjuik M, Agnamey P, Babiker A, et al. Amodiaquine-artesunate versus amodiaquine for uncomplicated Plasmodium falciparum malaria in African children: a randomised, multicentre trial. Lancet 2002; 359: 1365–72. Obonyo CO, Ochieng F, Taylor WR, et al. Artesunate plus sulfadoxine-pyrimethamine for uncomplicated malaria in Kenyan children: a randomized, double-blind, placebo-controlled trial. Trans R Soc Trop Med Hyg 2003; 97: 585–91. Nosten F, van Vugt M, Price R, et al. Effects of artesunatemefloquine combination on incidence of Plasmodium falciparum malaria and mefloquine resistance in western Thailand: a prospective study. Lancet 2000; 356: 297–302. Staedke SG, Mpimbaza A, Kamya MR, Nzarubara BK, Dorsey G, Rosenthal PJ. Combination treatments for uncomplicated falciparum malaria in Kampala, Uganda: randomised clinical trial. Lancet 2004; 364: 1950–57. Medicines for Malaria Venture. http://www.mmv.org (accessed Jan 23, 2005). Tran TH, Dolecek C, Pham PM, et al. Dihydroartemisininpiperaquine against multidrug-resistant Plasmodium falciparum malaria in Vietnam: randomised clinical trial. Lancet 2004; 363: 18–22. Alloueche A, Bailey W, Barton S, et al. Comparison of chlorproguanil-dapsone with sulfadoxine-pyrimethamine for the treatment of uncomplicated falciparum malaria in young African children: double-blind randomised controlled trial. Lancet 2004; 363: 1843–48. Mutabingwa T, Nzila A, Mberu E, et al. Chlorproguanil-dapsone for treatment of drug-resistant falciparum malaria in Tanzania. Lancet 2001; 358: 1218–23. World Health Organization. Assessment of the safety of artemisinin compounds in pregnancy. Geneva: WHO, 2003. Whitty CJ, Allan R, Wiseman V, et al. Averting a malaria disaster in Africa—where does the buck stop? Bull World Health Organ 2004; 82: 381–84. Dondorp AM, Newton PN, Mayxay M, et al. Fake antimalarials in Southeast Asia are a major impediment to malaria control: multinational cross-sectional survey on the prevalence of fake antimalarials. Trop Med Int Health 2004; 9: 1241–46. Vennerstrom JL, Arbe-Barnes S, Brun R, et al. Identification of an antimalarial synthetic trioxolane drug development candidate. Nature 2004; 430: 900–04. Arrow KJ, Panosian CB, Gelband H. Saving lives, buying time. Economics of malaria drugs in an age of resistance. Washington, DC: Institute of Medicine, National Academies Press, 2004.

1496

www.thelancet.com Vol 365 April 23, 2005

Seminar

86 87

88

89

90

91 92

93

94

95

96

97

98

99

100

101

102

103

104

World Health Organization. Scaling up home based care for malaria: from research to implementation. Geneva: WHO, 2004. Marsh VM, Mutemi WM, Willetts A, et al. Improving malaria home treatment by training drug retailers in rural Kenya. Trop Med Int Health 2004; 9: 451–60. Connor J, Rafter N, Rodgers A. Do fixed-dose combination pills or unit-of-use packaging improve adherence? A systematic review. Bull World Health Organ 2004; 82: 935–39. Barnes KI, Mwenechanya J, Tembo M, et al. Efficacy of rectal artesunate compared with parenteral quinine in initial treatment of moderately severe malaria in African children and adults: a randomised study. Lancet 2004; 363: 1598–605. Health Protection Agency. Malaria guidelines. http://www.hpa. org.uk/infections/topics_az/malaria/guidelines.htm (accessed Jan 23, 2005). Centers for Disease Control and Prevention. Malaria. http://www. cdc.gov/malaria (accessed Jan 23, 2005). Greenwood BM. The use of anti-malarial drugs to prevent malaria in the population of malaria-endemic areas. Am J Trop Med Hyg 2004; 70: 1–7. Schultz LJ, Steketee RW, Macheso A, et al. The efficacy of antimalarial regimens containing sulfadoxine-pyrimethamine and/or chloroquine in preventing peripheral and placental Plasmodium falciparum infection among pregnant women in Malawi. Am J Trop Med Hyg 1994; 51: 515–22. Shulman CE, Dorman EK, Cutts F, et al. Intermittent sulphadoxine- pyrimethamine to prevent severe anaemia secondary to malaria in pregnancy: a randomised placebo-controlled trial. Lancet 1999; 353: 632–36. Rogerson SJ, Chaluluka E, Kanjala M, et al. Intermittent sulphadoxine-pyrimethamine in pregnancy: effectiveness against malaria morbidity in Blantyre, Malawi, in 1997–99. Trans R Soc Trop Med Hyg 2000; 94: 549–53. Van Eijk AM, Ayisi JG, ter Kuile FO, et al. Effectiveness of intermittent preventive treatment with sulphadoxinepyrimethamine for control of malaria in pregnancy in western Kenya: a hospital-based study. Trop Med Int Health 2004; 9: 351–60. Schellenberg D, Menendez C, Kahigwa E, et al. Intermittent treatment for malaria and anaemia control at time of routine vaccinations in Tanzanian infants: a randomised, placebocontrolled trial. Lancet 2001; 357: 1471–77. Massaga JJ, Kitua AY, Lemnge MM, et al. Effect of intermittent treatment with amodiaquine on anaemia and malaria fever in infants in Tanzania: a randomised placebo-controlled trial. Lancet 2003; 361: 1853–60. Schellenberg D, Menendez C, Aponte JJ, et al. Intermittent preventive antimalarial treatment for Tanzanian infants: follow-up to age 2 years of a randomised, placebo-controlled trial. Lancet 2005; 365: 1481–83. Intermittent Preventive Treatment in Infants Consortium. Intermittent preventive treatment in infants (IPTi) with antimalarial drugs delivered through the expanded programme on immunization (EPI). http://www.ipti-malaria.org (accessed Jan 23, 2005). Cisse B, Sokhna CS, Alexander N, Trape J-F, Greenwood BM. A double blind, randomised placebo-controlled trial to measure the potential of sIPT with artesunate plus SP to reduce the malaria burden in Niakhar, rural Senegal. 53rd meeting of the American Society of Tropical Medicine and Hygiene, Miami, Nov 7–11, 2004, abstract 17. Dicko A, Sagara I, Sissoko MS, et al. Impact of intermittent preventive treatment with sulfadoxine-pyrimethamine targeting the transmission season on the incidence of clinical malaria in children of 6 months to 10 years in Kambila, Mali. 53rd meeting of the American Society of Tropical Medicine and Hygiene, Miami, Nov 7–11, 2004, abstract 18. Verhoeff H, West CE, Nzyuko SM, et al. Intermittent administration of iron and sulfadoxine-pyrimethamine to control anaemia in Kenyan children: a randomised controlled trial. Lancet 2002; 360: 908–14. Shankar AH, Genton B, Semba RD, et al. Effect of vitamin A supplementation on morbidity due to Plasmodium falciparum in young children in Papua New Guinea: a randomised trial. Lancet 1999; 354: 203–09.

105 Shankar AH, Genton B, Baisor M, et al. The influence of zinc supplementation on morbidity due to Plasmodium falciparum: a randomized trial in preschool children in Papua New Guinea. Am J Trop Med Hyg 2000; 62: 663–69. 106 Muller O, Becher H, van Zweeden AB, et al. Effect of zinc supplementation on malaria and other causes of morbidity in West African children: randomised double blind placebo controlled trial. BMJ 2001; 322: 1567–70. 107 Brey PT. Anopheles gambiae genome: perspectives for malaria control Mol Cells 2003; 15: 133–38. 108 Hallem EA, Fox N, Zwiebel J, Carlson JR. Olfaction: mosquito receptor for human-sweat odorant. Nature 2004; 427: 212–13. 109 Coleman PG, Alphey L. Editorial: genetic control of vector populations: an imminent prospect. Trop Med Int Health 2004; 9: 433–37. 110 Alphey L, Beard CB, Billingsley P, et al. Malaria control with genetically manipulated insect vectors. Science 2002; 298: 119–21. 111 Chen A, Rogan WJ. Nonmalarial infant deaths and DDT use for malaria control. Emerg Infect Dis 2003; 9: 960–64. 112 Mabaso ML, Sharp B, Lengeler C. Historical review of malarial control in southern Africa with emphasis on the use of indoor residual house-spraying. Trop Med Int Health 2004; 9: 846–56. 113 Lengeler C. Insecticide-treated bednets and curtains for preventing malaria. Cochrane Database Syst Rev 2004; 2: CD000363. 114 Phillips-Howard PA, Nahlen BJ, Kolczak MS, et al. Efficacy of permethrin-treated bed nets in the prevention of mortality in young children in an area of high perennial malaria transmission in western Kenya. Am J Trop Med Hyg 2003; 68 (suppl 4): 23–29. 115 Hawley WA, Phillips-Howard PA, ter Kuile FO, et al. Communitywide effects of permethrin-treated bed nets on child mortality and malaria morbidity in Western Kenya. Am J Trop Med Hyg 2003; 68 (suppl 4): 121–27. 116 Diallo DA, Cousens SN, Cuzin-Ouattera N, Nebie I, Ilboudo-Sanogo E, Esposito F. Child mortality in a West African population protected with insecticide-treated curtains for a period of up to 6 years. Bull World Health Organ 2004; 82: 85–91. 117 Binka FN, Hodgson A, Adjuik M, Smith T. Mortality in a sevenand-a-half year follow-up of a trial of insecticide-treated mosquito nets in Ghana. Trans R Soc Trop Med Hyg 2002; 96: 597–99. 118 World Health Organization. Report of the fourth meeting of the global collaboration for development of pesticides for public health. WHO 2004; WHO/CDS/WHOPES/GCDPP/2004·8. 119 Rowland M, Downey G, Rab A, et al. DEET mosquito repellent provides personal protection against malaria: a household randomized trial in an Afghan refugee camp in Pakistan. Trop Med Int Health 2004; 9: 335–42. 120 Brooke BD, Kloke G, Hunt RH, et al. Bioassay and biochemical analyses of insecticide resistance in southern African Anopheles funestus (Diptera: Culicidae). Bull Entomol Res 2001; 91: 265–72. 121 Zaim M, Guillet P. Alternative insecticides: an urgent need. Trends Parasitol 2002; 18: 161–63. 122 Monasch R, Reinisch A, Steketee R, et al. Child coverage with mosquito nets and malaria treatment from population-based surveys in African countries: a baseline for monitoring progress in roll back malaria. Am J Trop Med Hyg 2004; 71 (suppl 2): 232–38. 123 Armstrong Schellenberg JRM, Abdulla S, Nathan R, et al. Effect of large-scale social marketing of insecticide-treated nets on child survival in rural Tanzania. Lancet 2001; 357: 1241–47. 124 Curtis C, Maxwell C, Lemnge M, et al. Scaling-up coverage with insecticide-treated nets against malaria in Africa: who should pay? Lancet Infect Dis 2003; 3: 304–07. 125 Sachs JD, McArthur JW. The Millennium Project: a plan for meeting the Millennium Development Goals. Lancet 2005; 365: 347–53. 126 Moorthy VS, Good MF, Hill AVS. Malaria vaccine developments. Lancet 2004; 363: 150–56. 127 Polley SD, McRobert L, Sutherland CJ. Vaccination for vivax malaria: targeting the invaders. Trends Parasitol 2004; 20: 99–102. 128 Greenwood B, Alonso P. Malaria Vaccine Trials. Chem Immunol 2002; 80: 366–95. 129 Stoute JA, Slaoui M, Heppner DG, et al. A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. N Engl J Med 1997; 336: 86–91.

www.thelancet.com Vol 365 April 23, 2005

1497

Seminar

130 Kester KE, McKinney DA, Tornieporth N, et al. Efficacy of recombinant circumsporozoite protein vaccine regimens against experimental Plasmodium falciparum malaria. J Infect Dis 2001; 183: 640–47. 131 Bojang KA, Milligan PJ, Pinder M, et al. Efficacy of RTS,S/AS02 malaria vaccine against Plasmodium falciparum infection in semiimmune adult men in The Gambia: a randomised trial. Lancet 2001; 358: 1927–34. 132 Alonso PL, Sacarlal J, Aponte JJ, et al. Efficacy of the RTS,S/AS02A vaccine against Plasmodium falciparum infection and disease in young African children: randomised controlled trial. Lancet 2004; 364: 1411–20. 133 Schneider J, Gilbert SC, Blanchard TJ, et al. Enhanced immunogenicity for CD8+ T cell induction and complete protective efficacy of malaria DNA vaccination by boosting with modified vaccinia virus Ankara. Nat Med 1998; 4: 397–402. 134 Anderson RJ, Hannan CM, Gilbert SC, et al. Enhanced CD8+ T cell immune responses and protection elicited against Plasmodium berghei malaria by prime boost immunization regimens using a novel attenuated fowlpox virus. J Immunol 2004; 172: 3094–100. 135 Moorthy VS, Imoukhuede EB, Keating S, et al. Phase 1 evaluation of 3 highly immunogenic prime-boost regimens, including a 12-month reboosting vaccination, for malaria vaccination in Gambian men. J Infect Dis 2004; 189: 2213–19.

136 Moorthy VS, Imoukhuede EB, Milligan P, et al. A randomised, double-blind, controlled vaccine efficacy trial of DNA/MVA METRAP against malaria infection in Gambian adults. PLoS Med 2004; 1: e33. 137 World Health Organization. Portfolio of candidate malaria vaccines currently in development, October, 2004. http://www.who.int/ vaccine_research/documents/en/malaria_table.pdf (accessed Jan 23, 2005). 138 Pombo DJ, Lawrence G, Hirunpetcharat C, et al. Immunity to malaria after administration of ultra-low doses of red cells infected with Plasmodium falciparum. Lancet 2002; 360: 610–17. 139 Kaslow DC. Transmission-blocking vaccines. Chem Immunol 2002; 80: 287–307. 140 Malaria Vaccine Initiative. http://www.malariavaccine.org (accessed Jan 23, 2005). 141 The Global Fund. http://www.theglobalfund.org (accessed Jan 23, 2005). 142 Duke T. Clinical care for seriously ill children in district hospitals: a global public health issue. Lancet 2004; 363: 1922–23.

1498

www.thelancet.com Vol 365 April 23, 2005

Sponsor Documents

Or use your account on DocShare.tips

Hide

Forgot your password?

Or register your new account on DocShare.tips

Hide

Lost your password? Please enter your email address. You will receive a link to create a new password.

Back to log-in

Close