You are on page 1of 66

Malaria

Plasmodium falciparum ring-forms and gametocytes in human blood.

Malaria is a mosquito-borne infectious disease caused by a eukaryotic protist of the


genus Plasmodium. It is widespread in tropical and subtropical regions, including parts of
the Americas, Asia, and Africa. Each year, there are approximately 350–500 million
cases of malaria, killing between one and three million people, the majority of whom are
young children in Sub-Saharan Africa. Ninety percent of malaria-related deaths occur in
Sub-Saharan Africa. Malaria is commonly associated with poverty, but is also a cause of
poverty and a major hindrance to economic development.

Five species of the plasmodium parasite can infect humans; the most serious forms of the
disease are caused by Plasmodium falciparum. Malaria caused by Plasmodium vivax,
Plasmodium ovale and Plasmodium malariae causes milder disease in humans that is not
generally fatal. A fifth species, Plasmodium knowlesi, is a zoonosis that causes malaria
in macaques but can also infect humans.

Malaria is naturally transmitted by the bite of a female Anopheles mosquito. When a


mosquito bites an infected person, a small amount of blood is taken, which contains
malaria parasites. These develop within the mosquito and about one week later, when the
mosquito takes its next blood meal, the parasites are injected into the person being bitten

PDF created with pdfFactory Pro trial version www.pdffactory.com


with the mosquito's saliva. After a period of between 2 weeks and several months
(occasionally years) spent in the liver, the malaria parasites start to multiply within,
causing symptoms that include fever and headache. In severe cases the disease worsens
leading to coma, and death.

A wide variety of antimalarial drugs are available to treat malaria. In the last 5 years
treatment of P. falciparum infections in endemic countries has been transformed by the
use of combinations of drugs containing an artemisinin derivative. Severe malaria is
treated with intravenous or intramuscular quinine or, increasingly, the artemisinin
derivative artesunate. Several drugs are also available to prevent malaria in travellers to
malaria-endemic countries (prophylaxis). Resistance has developed to several
antimalarial drugs, most notably chloroquine.

Malaria transmission can be reduced by preventing mosquito bites with mosquito nets
and insect repellents, or by mosquito control measures such as spraying insecticides
inside houses and draining standing water where mosquitoes lay their eggs.

Although many are under development, the challenge of producing a widely available
vaccine that provides a high level of protection for a sustained period is still to be met

Signs and symptoms

Main symptoms of malaria.

Symptoms of malaria include fever, shivering, arthralgia (joint pain), vomiting,


anemia (caused by hemolysis), hemoglobinuria, retinal damage, and convulsions. The
classic symptom of malaria is cyclical occurrence of sudden coldness followed by rigor
and then fever and sweating lasting four to six hours, occurring every two days in P.
vivax and P. ovale infections, while every three for P. malariae. P. falciparum can have
recurrent fever every 36–48 hours or a less pronounced and almost continuous fever. For

PDF created with pdfFactory Pro trial version www.pdffactory.com


reasons that are poorly understood, but that may be related to high intracranial pressure,
children with malaria frequently exhibit abnormal posturing, a sign indicating severe
brain damage. Malaria has been found to cause cognitive impairments, especially in
children. It causes widespread anemia during a period of rapid brain development and
also direct brain damage. This neurologic damage results from cerebral malaria to which
children are more vulnerable. Cerebral malaria is associated with retinal whitening,
which may be a useful clinical sign in distinguishing malaria from other causes of fever.

Persistent in
Species Appearance Periodicity
liver?

Plasmodium vivax tertian yes

Plasmodium ovale tertian yes

Plasmodium
tertian no
falciparum

PDF created with pdfFactory Pro trial version www.pdffactory.com


Plasmodium malariae quartan no

Severe malaria is almost exclusively caused by P. falciparum infection and usually arises
6–14 days after infection. Consequences of severe malaria include coma and death if
untreated—young children and pregnant women are especially vulnerable. Splenomegaly
(enlarged spleen), severe headache, cerebral ischemia, hepatomegaly (enlarged liver),
hypoglycemia, and hemoglobinuria with renal failure may occur. Renal failure may cause
blackwater fever, where hemoglobin from lysed red blood cells leaks into the urine.
Severe malaria can progress extremely rapidly and cause death within hours or days. In
the most severe cases of the disease fatality rates can exceed 20%, even with intensive
care and treatment. In endemic areas, treatment is often less satisfactory and the overall
fatality rate for all cases of malaria can be as high as one in ten. Over the longer term,
developmental impairments have been documented in children who have suffered
episodes of severe malaria.

Chronic malaria is seen in both P. vivax and P. ovale, but not in P. falciparum. Here, the
disease can relapse months or years after exposure, due to the presence of latent parasites
in the liver. Describing a case of malaria as cured by observing the disappearance of
parasites from the bloodstream can, therefore, be deceptive. The longest incubation
period reported for a P. vivax infection is 30 years. Approximately one in five of P. vivax
malaria cases in temperate areas involve overwintering by hypnozoites (i.e., relapses
begin the year after the mosquito bite).

Causes

PDF created with pdfFactory Pro trial version www.pdffactory.com


A Plasmodium sporozoite traverses the cytoplasm of a mosquito midgut epithelial cell in
this false-color electron micrograph.

Malaria parasites

Malaria parasites are members of the genus Plasmodium (phylum Apicomplexa). In


humans malaria is caused by P. falciparum, P. malariae, P. ovale, P. vivax and P.
knowlesi. P. falciparum is the most common cause of infection and is responsible for
about 80% of all malaria cases, and is also responsible for about 90% of the deaths from
malaria. Parasitic Plasmodium species also infect birds, reptiles, monkeys, chimpanzees
and rodents. There have been documented human infections with several simian species
of malaria, namely P. knowlesi, P. inui, P. cynomolgi, P. simiovale, P. brazilianum, P.
schwetzi and P. simium; however, with the exception of P. knowlesi, these are mostly of
limited public health importance.

Mosquito vectors and the Plasmodium life cycle

The parasite's primary (definitive) hosts and transmission vectors are female mosquitoes
of the Anopheles genus, while humans and other vertebrates are secondary hosts. Young
mosquitoes first ingest the malaria parasite by feeding on an infected human carrier and
the infected Anopheles mosquitoes carry Plasmodium sporozoites in their salivary
glands. A mosquito becomes infected when it takes a blood meal from an infected
human. Once ingested, the parasite gametocytes taken up in the blood will further
differentiate into male or female gametes and then fuse in the mosquito gut. This
produces an ookinete that penetrates the gut lining and produces an oocyst in the gut wall.
When the oocyst ruptures, it releases sporozoites that migrate through the mosquito's
body to the salivary glands, where they are then ready to infect a new human host. This
type of transmission is occasionally referred to as anterior station transfer. The
sporozoites are injected into the skin, alongside saliva, when the mosquito takes a
subsequent blood meal.

Only female mosquitoes feed on blood, thus males do not transmit the disease. The
females of the Anopheles genus of mosquito prefer to feed at night. They usually start
searching for a meal at dusk, and will continue throughout the night until taking a meal.
Malaria parasites can also be transmitted by blood transfusions, although this is rare

PDF created with pdfFactory Pro trial version www.pdffactory.com


Pathogenesis

The life cycle of malaria parasites in the human body. A mosquito infects a person
by taking a blood meal.
First, sporozoites enter the bloodstream, and migrate to the liver.
They infect liver cells (hepatocytes), where they multiply into merozoites, rupture
the liver cells, and escape back into the bloodstream.
Then, the merozoites infect red blood cells, where they develop into ring forms, then
trophozoites (a feeding stage), then schizonts (a reproduction stage), then back into
merozoites.
Sexual forms called gametocytes are also produced, which, if taken up by a
mosquito, will infect the insect and continue the life cycle.

Malaria in humans develops via two phases: an exoerythrocytic and an erythrocytic


phase. The exoerythrocytic phase involves infection of the hepatic system, or liver,
whereas the erythrocytic phase involves infection of the erythrocytes, or red blood cells.
When an infected mosquito pierces a person's skin to take a blood meal, sporozoites in
the mosquito's saliva enter the bloodstream and migrate to the liver. Within 30 minutes of
being introduced into the human host, the sporozoites infect hepatocytes, multiplying
asexually and asymptomatically for a period of 6–15 days. Once in the liver, these
organisms differentiate to yield thousands of merozoites, which, following rupture of
their host cells, escape into the blood and infect red blood cells, thus beginning the

PDF created with pdfFactory Pro trial version www.pdffactory.com


erythrocytic stage of the life cycle. The parasite escapes from the liver undetected by
wrapping itself in the cell membrane of the infected host liver cell.

Within the red blood cells, the parasites multiply further, again asexually, periodically
breaking out of their hosts to invade fresh red blood cells. Several such amplification
cycles occur. Thus, classical descriptions of waves of fever arise from simultaneous
waves of merozoites escaping and infecting red blood cells.

Some P. vivax and P. ovale sporozoites do not immediately develop into exoerythrocytic-
phase merozoites, but instead produce hypnozoites that remain dormant for periods
ranging from several months (6–12 months is typical) to as long as three years. After a
period of dormancy, they reactivate and produce merozoites. Hypnozoites are responsible
for long incubation and late relapses in these two species of malaria.

The parasite is relatively protected from attack by the body's immune system because for
most of its human life cycle it resides within the liver and blood cells and is relatively
invisible to immune surveillance. However, circulating infected blood cells are destroyed
in the spleen. To avoid this fate, the P. falciparum parasite displays adhesive proteins on
the surface of the infected blood cells, causing the blood cells to stick to the walls of
small blood vessels, thereby sequestering the parasite from passage through the general
circulation and the spleen. This "stickiness" is the main factor giving rise to hemorrhagic
complications of malaria. High endothelial venules (the smallest branches of the
circulatory system) can be blocked by the attachment of masses of these infected red
blood cells. The blockage of these vessels causes symptoms such as in placental and
cerebral malaria. In cerebral malaria the sequestrated red blood cells can breach the blood
brain barrier possibly leading to coma.

Although the red blood cell surface adhesive proteins (called PfEMP1, for Plasmodium
falciparum erythrocyte membrane protein 1) are exposed to the immune system, they do
not serve as good immune targets because of their extreme diversity; there are at least 60
variations of the protein within a single parasite and effectively limitless versions within
parasite populations. The parasite switches between a broad repertoire of PfEMP1 surface
proteins, thus staying one step ahead of the pursuing immune system.

Some merozoites turn into male and female gametocytes. If a mosquito pierces the skin
of an infected person, it potentially picks up gametocytes within the blood. Fertilization
and sexual recombination of the parasite occurs in the mosquito's gut, thereby defining
the mosquito as the definitive host of the disease. New sporozoites develop and travel to
the mosquito's salivary gland, completing the cycle. Pregnant women are especially
attractive to the mosquitoes, and malaria in pregnant women is an important cause of
stillbirths, infant mortality and low birth weight, particularly in P. falciparum infection,
but also in other species infection, such as P. vivax.

PDF created with pdfFactory Pro trial version www.pdffactory.com


Diagnosis
Blood film

Blood smear from a P. falciparum culture (K1 strain). Several red blood cells have ring
stages inside them. Close to the center there is a schizont and on the left a trophozoite.

Since Charles Laveran first visualised the malaria parasite in blood in 1880, the mainstay
of malaria diagnosis has been the microscopic examination of blood.

Fever and septic shock are commonly misdiagnosed as severe malaria in Africa, leading
to a failure to treat other life-threatening illnesses. In malaria-endemic areas, parasitemia
does not ensure a diagnosis of severe malaria because parasitemia can be incidental to
other concurrent disease. Recent investigations suggest that malarial retinopathy is better
(collective sensitivity of 95% and specificity of 90%) than any other clinical or laboratory
feature in distinguishing malarial from non-malarial coma.

Although blood is the sample most frequently used to make a diagnosis, both saliva and
urine have been investigated as alternative, less invasive specimens.

Symptomatic diagnosis

Areas that cannot afford even simple laboratory diagnostic tests often use only a history
of subjective fever as the indication to treat for malaria. Using Giemsa-stained blood
smears from children in Malawi, one study showed that when clinical predictors (rectal
temperature, nailbed pallor, and splenomegaly) were used as treatment indications, rather
than using only a history of subjective fevers, a correct diagnosis increased from 21% to
41% of cases and unnecessary treatment for malaria was significantly decreased.

PDF created with pdfFactory Pro trial version www.pdffactory.com


Microscopic examination of blood films

For more details on individual parasites, see P. falciparum, P. vivax, P. ovale, P.


malariae.

The most economic, preferred, and reliable diagnosis of malaria is microscopic


examination of blood films because each of the four major parasite species has
distinguishing characteristics. Two sorts of blood film are traditionally used. Thin films
are similar to usual blood films and allow species identification because the parasite's
appearance is best preserved in this preparation. Thick films allow the microscopist to
screen a larger volume of blood and are about eleven times more sensitive than the thin
film, so picking up low levels of infection is easier on the thick film, but the appearance
of the parasite is much more distorted and therefore distinguishing between the different
species can be much more difficult. With the pros and cons of both thick and thin smears
taken into consideration, it is imperative to utilize both smears while attempting to make
a definitive diagnosis.

From the thick film, an experienced microscopist can detect parasite levels (or
parasitemia) down to as low as 0.0000001% of red blood cells. Diagnosis of species can
be difficult because the early trophozoites ("ring form") of all four species look identical
and it is never possible to diagnose species on the basis of a single ring form; species
identification is always based on several trophozoites.

Rapid antigen tests

OptiMAL-IT will reliably detect falciparum down to 0.01% parasitemia and non-
falciparum down to 0.1%. Paracheck-Pf will detect parasitemias down to 0.002% but
will not distinguish between falciparum and non-falciparum malaria. Parasite nucleic
acids are detected using polymerase chain reaction. This technique is more accurate than
microscopy. However, it is expensive, and requires a specialized laboratory. Moreover,
levels of parasitemia are not necessarily correlative with the progression of disease,
particularly when the parasite is able to adhere to blood vessel walls. Therefore more
sensitive, low-tech diagnosis tools need to be developed in order to detect low levels of
parasitaemia in the field.

Prevention

PDF created with pdfFactory Pro trial version www.pdffactory.com


Anopheles albimanus mosquito feeding on a human arm. This mosquito is a vector of
malaria and mosquito control is a very effective way of reducing the incidence of
malaria.

Methods used to prevent the spread of disease, or to protect individuals in areas where
malaria is endemic, include prophylactic drugs, mosquito eradication, and the prevention
of mosquito bites. The continued existence of malaria in an area requires a combination
of high human population density, high mosquito population density, and high rates of
transmission from humans to mosquitoes and from mosquitoes to humans. If any of these
is lowered sufficiently, the parasite will sooner or later disappear from that area, as
happened in North America, Europe and much of Middle East. However, unless the
parasite is eliminated from the whole world, it could become re-established if conditions
revert to a combination that favors the parasite's reproduction. Many countries are seeing
an increasing number of imported malaria cases due to extensive travel and migration.

The distribution of funding varies among countries. Countries with large populations do
not receive the same amount of support. The 34 countries that received a per capita
annual support of less than $1 included some of the poorest countries in Africa.

Brazil, Eritrea, India, and Vietnam have, unlike many other developing nations,
successfully reduced the malaria burden. Common success factors included conducive
country conditions, a targeted technical approach using a package of effective tools, data-
driven decision-making, active leadership at all levels of government, involvement of
communities, decentralized implementation and control of finances, skilled technical and
managerial capacity at national and sub-national levels, hands-on technical and
programmatic support from partner agencies, and sufficient and flexible financing.

Prophylactic drugs

Several drugs, most of which are also used for treatment of malaria, can be taken
preventively. Generally, these drugs are taken daily or weekly, at a lower dose than
would be used for treatment of a person who had actually contracted the disease. Use of
prophylactic drugs is seldom practical for full-time residents of malaria-endemic areas,
and their use is usually restricted to short-term visitors and travelers to malarial regions.
This is due to the cost of purchasing the drugs, negative side effects from long-term use,
and because some effective anti-malarial drugs are difficult to obtain outside of wealthy
nations.

Quinine was used starting in the 17th century as a prophylactic against malaria. The
development of more effective alternatives such as quinacrine, chloroquine, and
primaquine in the 20th century reduced the reliance on quinine. Today, quinine is still
used to treat chloroquine resistant Plasmodium falciparum, as well as severe and
cerebral stages of malaria, but is not generally used for prophylaxis.

Modern drugs used preventively include mefloquine (Lariam), doxycycline (available


generically), and the combination of atovaquone and proguanil hydrochloride

10

PDF created with pdfFactory Pro trial version www.pdffactory.com


(Malarone). The choice of which drug to use depends on which drugs the parasites in the
area are resistant to, as well as side-effects and other considerations. The prophylactic
effect does not begin immediately upon starting taking the drugs, so people temporarily
visiting malaria-endemic areas usually begin taking the drugs one to two weeks before
arriving and must continue taking them for 4 weeks after leaving (with the exception of
atovaquone proguanil that only needs be started 2 days prior and continued for 7 days
afterwards).

The use of prophylactic drugs where malaria-bearing mosquitoes are present may
encourage the development of partial immunity.

Treatment
Active malaria infection with P. falciparum is a medical emergency requiring
hospitalization. Infection with P. vivax, P. ovale or P. malariae can often be treated on an
outpatient basis. Treatment of malaria involves supportive measures as well as specific
antimalarial drugs. When properly treated, someone with malaria can expect a complete
recovery.

Counterfeit drugs

Sophisticated counterfeits have been found in several Asian countries such as Cambodia,
China, Indonesia, Laos, Thailand, Vietnam and are an important cause of avoidable death
in those countries. WHO have said that studies indicate that up to 40% of artesunate
based malaria medications are counterfeit, especially in the Greater Mekong region and
have established a rapid alert system to enable information about counterfeit drugs to be
rapidly reported to the relevant authorities in participating countries. There is no reliable
way for doctors or lay people to detect counterfeit drugs without help from a laboratory.
Companies are attempting to combat the persistence of counterfeit drugs by using new
technology to provide security from source to distribution.

Epidemiology

11

PDF created with pdfFactory Pro trial version www.pdffactory.com


Countries which have regions where malaria is endemic as of 2003 (coloured yellow
Countries in green are free of indigenous cases of malaria in all areas.

Disability-adjusted life year for malaria per 100,000 inhabitants in 2002.


no data ≤10 10-50 50-100 100-250 250-500 500-1000 1000-1500 1500-
2000 2000-2500 2500-3000 3000-3500 ≥3500

Malaria causes about 250 million cases of fever and approximately one million deaths
annually. The vast majority of cases occur in children under 5 years old; pregnant women
are also especially vulnerable. Despite efforts to reduce transmission and increase
treatment, there has been little change in which areas are at risk of this disease since
1992. Indeed, if the prevalence of malaria stays on its present upwards course, the death
rate could double in the next twenty years. Precise statistics are unknown because many
cases occur in rural areas where people do not have access to hospitals or the means to
afford health care. As a consequence, the majority of cases are undocumented.

Although co-infection with HIV and malaria does cause increased mortality, this is less
of a problem than with HIV/tuberculosis co-infection, due to the two diseases usually
attacking different age-ranges, with malaria being most common in the young and active
tuberculosis most common in the old. Although HIV/malaria co-infection produces less
severe symptoms than the interaction between HIV and TB, HIV and malaria do
contribute to each other's spread. This effect comes from malaria increasing viral load
and HIV infection increasing a person's susceptibility to malaria infection.

Malaria is presently endemic in a broad band around the equator, in areas of the
Americas, many parts of Asia, and much of Africa; however, it is in sub-Saharan Africa
where 85– 90% of malaria fatalities occur. The geographic distribution of malaria within
large regions is complex, and malaria-afflicted and malaria-free areas are often found
close to each other. In drier areas, outbreaks of malaria can be predicted with reasonable
accuracy by mapping rainfall. Malaria is more common in rural areas than in cities; this is

12

PDF created with pdfFactory Pro trial version www.pdffactory.com


in contrast to dengue fever where urban areas present the greater risk. For example, the
cities of Vietnam, Laos and Cambodia are essentially malaria-free, but the disease is
present in many rural regions. By contrast, in Africa malaria is present in both rural and
urban areas, though the risk is lower in the larger cities. The global endemic levels of
malaria have not been mapped since the 1960s. However, the Wellcome Trust, UK, has
funded the Malaria Atlas Project to rectify this, providing a more contemporary and
robust means with which to assess current and future malaria disease burden.

History
Malaria has infected humans for over 50,000 years, and Plasmodium may have been a
human pathogen for the entire history of the species. Close relatives of the human malaria
parasites remain common in chimpanzees. References to the unique periodic fevers of
malaria are found throughout recorded history, beginning in 2700 BC in China. The term
malaria originates from Medieval Italian: mala aria—"bad air"; and the disease was
formerly called ague or marsh fever due to its association with swamps and marshland.
Malaria was once common in most of Europe and North America, where it is no longer
endemic, though imported cases do occur.

Scientific studies on malaria made their first significant advance in 1880, when a French
army doctor working in the military hospital of Constantine in Algeria named Charles
Louis Alphonse Laveran observed parasites for the first time, inside the red blood cells of
people suffering from malaria. He, therefore, proposed that malaria is caused by this
organism, the first time a protist was identified as causing disease. For this and later
discoveries, he was awarded the 1907 Nobel Prize for Physiology or Medicine. The
malarial parasite was called Plasmodium by the Italian scientists Ettore Marchiafava and
Angelo Celli. A year later, Carlos Finlay, a Cuban doctor treating patients with yellow
fever in Havana, provided strong evidence that mosquitoes were transmitting disease to
and from humans. This work followed earlier suggestions by Josiah C. Nott, and work by
Patrick Manson on the transmission of filariasis.

However, it was Britain's Sir Ronald Ross working in the Presidency General Hospital in
Calcutta who finally proved in 1898 that malaria is transmitted by mosquitoes. He did
this by showing that certain mosquito species transmit malaria to birds and isolating
malaria parasites from the salivary glands of mosquitoes that had fed on infected birds.
For this work Ross received the 1902 Nobel Prize in Medicine. After resigning from the
Indian Medical Service, Ross worked at the newly-established Liverpool School of
Tropical Medicine and directed malaria-control efforts in Egypt, Panama, Greece and
Mauritius. The findings of Finlay and Ross were later confirmed by a medical board
headed by Walter Reed in 1900, and its recommendations implemented by William C.
Gorgas in the health measures undertaken during construction of the Panama Canal. This
public-health work saved the lives of thousands of workers and helped develop the
methods used in future public-health campaigns against this disease.

The first effective treatment for malaria came from the bark of cinchona tree, which
contains quinine. This tree grows on the slopes of the Andes, mainly in Peru. A tincture

13

PDF created with pdfFactory Pro trial version www.pdffactory.com


made of this natural product was used by the inhabitants of Peru to control malaria, and
the Jesuits introduced this practice to Europe during the 1640s, where it was rapidly
accepted. However, it was not until 1820 that the active ingredient, quinine, was
extracted from the bark, isolated and named by the French chemists Pierre Joseph
Pelletier and Joseph Bienaimé Caventou.

In the early 20th century, before antibiotics became available, Julius Wagner-Jauregg
discovered that patients with syphilis could be treated by intentionally infecting them
with malaria; the resulting fever would kill the syphilis spirochetes, and quinine would
then be administered to control the malaria. Although some patients died from malaria,
this was considered preferable to the almost-certain death from syphilis.

Although the blood stage and mosquito stages of the malaria life cycle were identified in
the 19th and early 20th centuries, it was not until the 1980s that the latent liver form of
the parasite was observed. The discovery of this latent form of the parasite finally
explained why people could appear to be cured of malaria but still relapse years after the
parasite had disappeared from their bloodstreams.

ANTIMALARIAL DRUGS

A thin-film Giemsa stained micrograph of ring-forms, and gametocytes of Plasmodium


falciparum. (CDC)

Antimalarial drugs are designed to prevent or cure malaria.

Uses of antimalarial drugs


• Treatment of malaria in individuals with suspected or confirmed infection
• Prevention of infection in individuals visiting a malaria-endemic region who have
no immunity (prophylaxis)
• Routine Intermittent treatment of certain groups in endemic regions

14

PDF created with pdfFactory Pro trial version www.pdffactory.com


• Some antimalarial agents, particularly chloroquine and hydroxychloroquine, are
also used in the treatment of rheumatoid arthritis and lupus associated arthritis.

Some agents are used for more than one application. It is therefore more practical to
group antimalarials by chemical structure since this is associated with important
properties of each drug, such as mechanism of action.

Quinine and related agents

Quinine has a long history stretching from Peru, and the discovery of the cinchona tree,
and the potential uses of its bark, to the current day and a collection of derivatives that are
still frequently used in the prevention and treatment of malaria. Quinine is an alkaloid
that acts as a blood schizonticidal and weak gametocide against Plasmodium vivax and
Plasmodium malariae. As an alkaloid, it is accumulated in the food vacuoles of
Plasmodium species, especially Plasmodium falciparum. It acts by inhibiting the
hemozoin biocrystallization, thus facilitating an aggregation of cytotoxic heme. Quinine
is less effective and more toxic as a blood schizonticidal agent than chloroquine; however
it is still very effective and widely used in the treatment of acute cases of severe P.
falciparum. It is especially useful in areas where there is known to be a high level of
resistance to chloroquine, mefloquine and sulfa drug combinations with pyrimethamine.
Quinine is also used in post-exposure treatment of individuals returning from an area
where malaria is endemic.

The treatment regimen of quinine is complex and is determined largely by the parasite's
level of resistance and the reason for drug therapy (i.e. acute treatment or prophylaxis).
The World Health Organization recommendation for quinine is 8 mg/kg three times daily
for 3 days in areas where the level of adherence is questionable and for 7 days where
parasites are sensitive to quinine. In areas where there is an increased level of resistance
to quinine 8 mg/kg three times daily for 7 days is recommended, combined with
doxycycline, tetracycline or clindamycin. Doses can be given by oral, intravenous or
intramuscular routes. The recommended method depends on the urgency of treatment and
the available resources (i.e. sterilised needles for IV or IM injections).

Use of quinine is characterised by a frequently experienced syndrome called cinchonism.


Tinnitus (a hearing impairment), rashes, vertigo, nausea, vomiting and abdominal pain
are the most common symptoms. Neurological effects are experienced in some cases due
to the drug's neurotoxic properties. These actions are mediated through the interactions of
Quinine causing a decrease in the excitability of the motor neuron end plates. This often
results in functional impairment of the eight cranial nerve; resulting in confusion,
delirium and coma. Quinine can cause hypoglycaemia through its action of stimulating
insulin secretion, this occurs in therapeutic doses and therefore it is advised that glucose
levels are monitored in all patients every 4–6 hours. This effect can be exaggerated in
pregnancy and therefore additional care in administering and monitoring the dosage is
essential. Repeated or over-dosage can result in renal failure and death through
depression of the respiratory system.

15

PDF created with pdfFactory Pro trial version www.pdffactory.com


Quinimax and quinidine are the two most commonly used alkaloids related to quinine, in
the treatment or prevention of malaria. Quinimax is a combination of four alkaloids
(quinine, quinidine, cinchoine and cinchonidine). This combination has been shown in
several studies to be more effective than quinine, supposedly due to a synergistic action
between the four cinchona derivatives. Quinidine is a direct derivative of quinine. It is a
distereoisomer, thus having similar anti-malarial properties to the parent compound.
Quinidine is recommended only for the treatment of severe cases of malaria.

Chloroquine

Chloroquine was until recently the most widely used anti-malarial. It was the original
prototype from which most other methods of treatment are derived. It is also the least
expensive, best tested and safest of all available drugs. The emergence of drug resistant
parasitic strains is rapidly decreasing its effectiveness; however it is still the first-line
drug of choice in most sub-Saharan African countries. It is now suggested that it is used
in combination with other antimalarial drugs to extend its effective usage.

Chloroquine is a 4-aminoquinolone compound with a complicated and still unclear


mechanism of action. It is believed to reach high concentrations in the vacuoles of the
parasite, which, due to its alkaline nature, raises the internal pH. It controls the
conversion of toxic heme to hemozoin by inhibiting the biocrystallization of hemozoin
thus poisoning the parasite through excess levels of toxicity. Other potential mechanisms
through which it may act include interfering with the biosynthesis of parasitic nucleic
acids, the formation of a chloroquine-haem or chloroquine-DNA complex. The most
significant level of activity found is against all forms of the schizonts (with the obvious
exception of chloroquine-resistant P. falciparum and P. vivax strains) and the
gametocytes of P. vivax, P. malariae, P. ovale as well as the immature gametocytes of P.
falciparum. Chloroquine also has a significant anti-pyretic and anti-inflammatory effect
when used to treat P. vivax infections, thus it may still remain useful even when
resistance is more widespread. According to a report on the Science and Development
Network website's sub-Saharan Africa section, there is very little drug resistance among
children infected with malaria on the island of Madagascar, but what drug resistance
there is, exists against chloroquinine.

A slightly different drug called nivaquine or chloroquine phosphate has also been
invented. Popular drugs that make use of this compound are Chloroquine FNA, Resochin
and Dawaquin.

Route of administration: It can be given orally, or intramuscularly.

Formulation/Strength: Tablet contains 250mg of chloroquine


diphosphate, corresponding to 150mg of chloroquine base.
Ampoule. Chloroquine injection, 40mg/ml.
Chloroqine injection: (I.M.I.)

- 40mg/ml
- I.M. injection only. (NEVER I.V.) For more serious cases, i.e. if the patient is vomiting, fitting,
not fully conscious, temp. over 39.5 degrees or severe diarrhea.

16

PDF created with pdfFactory Pro trial version www.pdffactory.com


- 0.1ml/kg given I.M.I. and repeated 6 hours later. Repeat 0.1ml/kg daily for up to 2 days if still
unconscious, but change to oral treatment as soon as possible.

Small adult: 5ml


(Under 50kg)

Large adult. 7ml


(Over 50kg)

Children: Give 0.1ml/kg I.M.I.

Weight (kg) Dose per ml (I.M.I.)

3–5 ¼
5–7 ¼
8–9 ¾
10 – 14 1
15 – 19 1½
20 – 24 2
25 – 29 2½
30 - 39 3

Change to amodiaquine or chloroquine tablet as soon as possible

Children and adults should receive 25 mg of chloroquine per kg given over 3 days. A
pharmacokinetically superior regime, recommended by the WHO, involves giving an
initial dose of 10 mg/kg followed 6–8 hours later by 5 mg/kg, then 5 mg/kg on the
following 2 days. For chemoprophylaxis: 5 mg/kg/week (single dose) or 10 mg/kg/week
divided into 6 daily doses is advised. It should be noted that chloroquine is only
recommended as a prophylactic drug in regions only affected by P. vivax and sensitive P.
falciparum strains. Chloroquine has been used in the treatment of malaria for many years
and no abortifacient or teratogenic effects have been reported during this time, therefore
it is considered very safe to use during pregnancy. However, itching can occur at
intolerable level.

Amodiaquine

Amodiaquine is a 4-aminoquinolone anti-malarial drug similar in structure and


mechanism of action to Chloroquine. It is most frequently used in combination with
Chloroquine, but is also very effective when used alone. It is thought to be more effective
in clearing parasites in uncomplicated malarial than Chloroquine, thus leading to a faster
rate of recovery. However, some fatal adverse effects of the drug were noted during the
1980s, thus reducing its usage in chemoprophylaxis. The WHO's most recent advice on
the subject still maintains that the drug should be used when the potential risk of not
treating an infection outweighs the risk of developing side effects. It has also been
suggested that it is particularly effective, and less toxic than other combination treatments
in HIV positive patients.

17

PDF created with pdfFactory Pro trial version www.pdffactory.com


Dosage

Weight (kg) Dose in tab/mg

3 – 5.9 ½ (50mg)
6 – 9.9 1 (100mg)
10 – 14.9 1½ (150mg)
15 – 18.9 2 (200mg)
19 & over give chloroquine

The drug should be given in doses between 25 mg/kg and 35 mg/kg over 3 days in a
similar method to that used in Chloroquine administration. Adverse reactions are
generally similar in severity and type to that seen in Chloroquine treatment. In addition,
bradycardia, itching, nausea, vomiting and some abdominal pain have been recorded.
Some blood and hepatic disorders have also been seen in a small number of patients.

Pyrimethamine

Pyrimethamine is used in the treatment of uncomplicated malaria. It is particularly useful


in cases of chloroquine-resistant P. Falciparum strains when combined with
Sulphadoxine. It acts by inhibiting dihydrofolate reductase in the parasite thus preventing
the biosynthesis of purines and pyrimidines. Therefore halting the processes of DNA
synthesis, cell division and reproduction. It acts primarily on the schizonts during the
hepatic and erythrocytic phases.

Sulphadoxine

The action of Sulphadoxine is focused on inhibiting the use of para-aminobenzoic acid


during the synthesis of dihydropteroic acid. When combined with Pyrimethamine the two
key stages in DNA synthesis in the plasmodia are prevented. It also acts on the schizonts
during the hepatic and erythrocytic phases. It is mainly used for treating P. falciparum
infections and is less active against other Plasmodium strains. However usage is
restricted due to the long half life of the combination which exerts a potentially large
selection pressure on the parasite hence encouraging the possibility of resistance
developing. This combination is not recommended for chemoprophylaxis because of the
severe skin reactions commonly experienced. However it is used frequently for clinical
episodes of the disease.

Proguanil

Proguanil (Chloroguanadine) is a biguanide; a synthetic derivative of pyrimidine. It was


developed in 1945 by a British Antimalarial research group. It has many mechanisms of
action but primarily is mediated through conversion to the active metabolite cycloguanil
pamoate. This inhibits the malarial dihydrofolate reductase enzyme. Its most prominent
effect is on the primary tissue stages of P. falciparum, P. vivax and P. ovale. It has no

18

PDF created with pdfFactory Pro trial version www.pdffactory.com


known effect against hypnozoites therefore is not used in the prevention of relapse. It has
a weak blood schizonticidal activity and is not recommended for therapy of acute
infection. However it is useful in prophylaxis when combined with Atovaquone or
chloroquine (in areas where there is no chloroquine resistance). 3 mg/kg is the advised
dosage per day, (hence approximate adult dosage is 200 mg). The pharmacokinetic
profile of the drugs indicates that a half dose, twice daily maintains the plasma levels
with a greater level of consistency, thus giving a greater level of protection. It should be
noted that the Proguanil- Chloroquine combination does not provide effective protection
against resistant strains of P. falciparum. There are very few side effects to Proguanil,
with slight hair loss and mouth ulcers being occasionally reported following prophylactic
use.

Mefloquine

Mefloquine was developed during the Vietnam War and is chemically related to quinine.
It was developed to protect American troops against multi-drug resistant P. falciparum. It
is a very potent blood schizonticide with a long half-life. It is thought to act by forming
toxic heme complexes that damage parasitic food vacuoles. It is now used solely for the
prevention of resistant strains of P. falciparum despite being effective against P. vivax, P.
ovale and P. marlariae. Mefloquine is effective in prophylaxis and for acute therapy. It is
now strictly used for resistant strains (and is usually combined with Artesunate).
Chloroquine/Proguanil or sufha drug-pyrimethamine combinations should be used in all
other Plasmodia infections.

The major commercial manufacturer of mefloquine-based malaria treatment is Roche


Pharmaceuticals, which markets the drug under the trade name "Lariam". Lariam is fairly
expensive at around 3 € per tablet (pricing of the year 2000).

A dose of 15–25 mg/kg is recommended, depending on the prevalence of Mefloquine


resistance. The increased dosage is associated with a much greater level of intolerance,
most noticeably in young children; with the drug inducing vomiting and oesophagitis.
The effects during pregnancy are unknown, although it has been linked with an increased
number of stillbirths. It is not recommended for use during the first trimester, although
considered safe during the second and third trimesters. Mefloquine frequently produces
side effects, including nausea, vomiting, diarrhea, abdominal pain and dizziness. Several
associations with neurological events have been made, namely affective and anxiety
disorders, hallucinations, sleep disturbances, psychosis, toxic encephalopathy,
convulsions and delirium. Cardiovascular effects have been recorded with bradycardia
and sinus arrhythmia being consistently recorded in 68% of patients treated with
Mefloquine (in one hospital-based study).

Mefloquine can only be taken for a period up to 6 months (due to side effects, ...). After
this, other drugs (such as those based on paludrine/nivaquine) again need to be taken.

19

PDF created with pdfFactory Pro trial version www.pdffactory.com


Atovaquone

Atovaquone is only available in combination with Proguanil under the name Malarone,
albeit at a price higher than Lariam. It is most commonly used in prophylaxis by
travellers.

Primaquine

Primaquine is a highly active 8-aminoquinolone that is used in treating all types of


malaria infection. It is most effective against gametocytes but also acts on hypnozoites,
blood schizonticytes and the dormant plasmodia in P. vivax and P. ovale. It is the only
known drug to cure both relapsing malaria infections and acute cases. The mechanism of
action is not fully understood but it is thought to mediate some effect through creating
oxygen free radicals that interfere with the plasmodial electron transport chain during
respiration.

Formulation/Strength: Tablet – 7.5mg

Route of administration: Administered orally.

Dosage:

Adult. For single dose on the first day of treatment for P. falciparum.

- Small adult 4 tablet (30mg)


(Under 50kg)

- Large adult 6 tablet (45mg)


(Over 50kg)

Children. For single dose on the first day of treatment.

Weight (kg) Dose in tab./mg

3 – 5.9 -
6 – 9.9 ½ (3.75mg)
10 – 14.9 1 (7.5mg)
15 – 18.9 1½ (11.25mg)
19 – 27.9 2 (15mg)
28 – 36.9 3 (22.5mg)
37 – 49.9 4 (30mg)

For the prevention of relapse in P. vivax and P. ovale 0.15 mg/kg should be given for 14
days. As a gametocytocidal drug in P. falciparum infections a single dose of 0.75 mg/kg
repeated 7 days later is sufficient. This treatment method is only used in conjunction with
another effective blood schizonticidal drug. There are few significant side effects
although is has been shown that Primaquine may cause anorexia, nausea, vomiting,
cramps, chest weakness, anaemia, some suppression of myeloid activity and abdominal
pains. In cases of over-dosage granulocytopenia may occur.

20

PDF created with pdfFactory Pro trial version www.pdffactory.com


Artemesinin and derivatives

Artemesinin is a Chinese herb (Qinghaosu) that has been used in the treatment of fevers
for over 1,000 years, thus predating the use of Quinine in the western world. It is derived
from the plant Artemisia annua, with the first documentation as a successful therapeutic
agent in the treatment of malaria is in 340 AD by Ge Hong in his book Zhou Hou Bei Ji
Fang (A Handbook of Prescriptions for Emergencies). The active compound was isolated
first in 1971 and named Artemsinin. It is a sesquiterpene lactone with a chemically rare
peroxide bridge linkage. It is this that is thought to be responsible for the majority of its
anti-malarial action, although the target within the parasite remains controversial. At
present it is strictly controlled under WHO guidelines as it has proven to be effective
against all forms of multi-drug resistant P. falciparum, thus every care is taken to ensure
compliance and adherence together with other behaviours associated with the
development of resistance. It is also only given in combination with other anti-malarials.

• Artemisinin has a very rapid action and the vast majority of acute patients
treated show significant improvement within 1–3 days of receiving treatment. It
has demonstrated the fastest clearance of all anti-malarials currently used and acts
primarily on the trophozite phase, thus preventing progression of the disease.
Semi-synthetic artemisinin derivatives (e.g. artesunate, artemether) are easier to
use than the parent compound and are converted rapidly once in the body to the
active compound dihydroartemesinin. On the first day of treatment 20 mg/kg
should be given, this dose is then reduced to 10 mg/kg per day for the 6 following
days. Few side effects are associated with artemesinin use. However, headaches,
nausea, vomiting, abnormal bleeding, dark urine, itching and some drug fever
have been reported by a small number of patients. Some cardiac changes were
reported during a clinical trial, notably non specific ST changes and a first degree
atrioventricular block (these disappeared when the patients recovered from the
malarial fever).

• Artemether is a methyl ether derivative of Dihydroartemesinin. It is similar to


Artemesinin in mode of action but demonstrates a reduced ability as a
hypnozoiticidal compound, instead acting more significantly to decrease
gametocyte carriage. Similar restrictions are in place, as with Artemesinin, to
prevent the development of resistance, therefore it is only used in combination
therapy for severe acute cases of drug-resistant P. falciparum. It should be
administered in a 7 day course with 4 mg/kg given per day for 3 days, followed
by 1.6 mg/kg for 3 days. Side effects of the drug are few but include potential
neurotoxicity developing if high doses are given.
• Artesunate is a hemisuccinate derivative of the active metabolite
Dihydroartemisin. Currently it is the most frequently used of all the Artemesinin-
type drugs. Its only effect is mediated through a reduction in the gametocyte
transmission. It is used in combination therapy and is effective in cases of
uncomplicated P. falciparum. The dosage recommended by the WHO is a 5 or 7
day course (depending on the predicted adherence level) of 4 mg/kg for 3 days
(usually given in combination with Mefloquine) followed by 2 mg/kg for the

21

PDF created with pdfFactory Pro trial version www.pdffactory.com


remaining 2 or 4 days. In large studies carried out on over 10,000 patients in
Thailand no adverse effects have been shown.
• Dihydroartemisinin is the active metabolite to which Artemesinin is reduced.
It is the most effective Artemesinin compound and the least stable. It has a strong
blood schizonticidal action and reduces gametocyte transmission. It is used for
therapeutic treatment of cases of resistant and uncomplicated P. falciparum.
4 mg/kg doses are recommended on the first day of therapy followed by 2 mg/kg
for 6 days. As with Artesunate, no side effects to treatment have thus far been
recorded.
• Arteether is an ethyl ether derivative of Dihydroartemisinin. It is used in
combination therapy for cases of uncomplicated resistant P. falciparum. The
recommended dosage is 150 mg/kg per day for 3 days given by IM injections.
With the exception of a small number of cases demonstrating neurotoxicity
following parenteral administration no side effects have been recorded.

Halofantrine

Halofantrine is a relatively new drug developed by the Walter Reed Army Institute of
Research in the 1960s. It is a phenanthrene methanol, chemically related to Quinine and
acts acting as a blood schizonticide effective against all plasmodium parasites. Its
mechanism of action is similar to other anti-malarials. Cytotoxic complexes are formed
with ferritoporphyrin XI that cause plasmodial membrane damage. Despite being
effective against drug resistant parasites, Halofantrine is not commonly used in the
treatment (prophylactic or therapeutic) of malaria due to its high cost. It has very variable
bioavailability and has been shown to have potentially high levels of cardiotoxicity. It is
still a useful drug and can be used in patients that are known to be free of heart disease
and are suffering from severe and resistant forms of acute malaria. A popular drug based
on halofantrine is Halfan. The level of governmental control and the prescription-only
basis on which it can be used contributes to the cost, thus Halofantrine is not frequently
used.

A dose of 8 mg/kg of Halofantrine is advised to be given in three doses at six hour


intervals for the duration of the clinical episode. It is not recommended for children under
10 kg despite data supporting the use and demonstrating that it is well tolerated. The most
frequently experienced side-effects include nausea, abdominal pain, diarrhoea, and itch.
Severe ventricular dysrhythmias, occasionally causing death are seen when high doses
are administered. This is due to prolongation of the QTc interval. Halofantrine is not
recommended for use in pregnancy and lactation, in small children, or in patients that
have taken Mefloquine previously. Lumefantrine is a relative of halofantrine that is used
in some combination antimalarial regimens.

Doxycycline

Probably one of the more prevalent antimalarial drugs prescribed, due to its relative
effectiveness and cheapness, Doxycycline is a Tetracycline compound derived from
Oxytetracycline. The tetracyclines were one of the earliest groups of antibiotics to be

22

PDF created with pdfFactory Pro trial version www.pdffactory.com


developed and are still used widely in many types of infection. It is a bacteriostatic agent
that acts to inhibit the process of protein synthesis by binding to the 30S ribosomal
subunit thus preventing the 50s and 30s units from bonding. Doxycycline is used
primarily for chemoprophylaxis in areas where chloroquine resistance exists. It can also
be used in combination with quinine to treat resistant cases of P. falciparum but has a
very slow action in acute malaria, and should not be used as monotherapy.

When treating acute cases and given in combination with Quinine; 100 mg/kg of
Doxycycline should be given per day for 7 days. In prophylactic therapy, 100 mg (adult
dose) of Doxycycline should be given every day during exposure to malaria.

The most commonly experienced side effects are permanent enamel hypoplasia, transient
depression of bone growth, gastrointestinal disturbances and some increased levels of
photosensitivity. Due to its effect of bone and tooth growth it is not used in children
under 8, pregnant or lactating women and those with a known hepatic dysfunction.

Tetracycline is only used in combination for the treatment of acute cases of P.Falciparum
infections. This is due to its slow onset. Unlike Doxycycline it is not used in
chemoprophylaxis. For Tetracycline, 250 mg is the recommended adult dosage (it should
not be used in children) for 5 or 7 days depending on the level of adherence and
compliance expected. Oesophageal ulceration, gastrointestinal upset and interferences
with the process of ossification and depression of bone growth are known to occur. The
majority of side effects associated with Doxycycline are also experienced.

Clindamycin

Clindamycin is a derivative of Lincomycin, with a slow action against blood


schizonticides. It is only used in combination with Quinine in the treatment of acute cases
of resistant P. falciparum infections and not as a prophylactic. Being more expensive and
toxic than the other antibiotic alternatives, it is used only in cases where the Tetracyclines
are contraindicated (for example in children).

Clindamycin should be given in conjunction with Quinine as a 300 mg dose (in adults)
four times a day for 5 days. The only side effects recorded in patients taking Clindamycin
are nausea, vomiting and abdominal pains and cramps. However these can be alleviated
by consuming large quantities of water and food when taking the drug.
Pseudomembranous colitis (caused by Clostridium difficile) has also developed in
some patients; this condition may be fatal in a small number of cases.

Resistance to antimalarials

Antimalarial resistance is common.

Anti-malarial drug resistance has been defined as: "the ability of a parasite to survive
and/or multiply despite the administration and absorption of a drug given in doses equal
to or higher than those usually recommended but within tolerance of the subject. The

23

PDF created with pdfFactory Pro trial version www.pdffactory.com


drug in question must gain access to the parasite or the infected red blood cell for the
duration of the time necessary for its normal action." In most instances this refers to
parasites that remaining following on from an observed treatment. Thus excluding all
cases where anti-malarial prophylaxis has failed. In order for a case to be defined as
resistant, the patient under question must have received a known and observed anti-
malarial therapy whilst the blood drug and metabolite concentrations are monitored
concurrently. The techniques used to demonstrate this are: in vivo, in vitro, animal model
testing and the most recently developed molecular techniques.

Drug resistant parasites are often used to explain malaria treatment failure. However, they
are two potentially very different clinical scenarios. The failure to clear parasitemia and
recover from an acute clinical episode when a suitable treatment has been given and anti-
malarial resistance in its true form. Drug resistance may lead to treatment failure, but
treatment failure is not necessarily caused by drug resistance despite assisting with its
development. A multitude of factors can be involved in the processes including problems
with non-compliance and adherence, poor drug quality, interactions with other
pharmaceuticals, poor absorption, misdiagnosis and incorrect doses being given. The
majority of these factors also contribute to the development of drug resistance.

The generation of resistance can be complicated and varies between plasmodium species.
It is generally accepted to be initiated primarily through a spontaneous mutation that
provides some evolutionary benefit, thus giving an anti-malarial used a reduced level of
sensitivity. This can be caused by a single point mutation or multiple mutations. In most
instances a mutation will be fatal for the parasite or the drug pressure will remove
parasites that remain susceptible, however some resistant parasites will survive.
Resistance can become firmly established within a parasite population, existing for long
periods of time.

The first type of resistance to be acknowledged was to Chloroquine in Thailand in 1957.


The biological mechanism behind this resistance was subsequently discovered to be
related to the development of an efflux mechanism that expels Chloroquine from the
parasite before the level required to effectively inhibit the process of haem
polymerization (that is necessary to prevent build up of the toxic by products formed by
haemoglobin digestion). This theory has been supported by evidence showing that
resistance can be effectively reversed on the addition of substances which halt the efflux.
The resistance of other quinolone anti-malarials such as amiodiaquine, mefloquine,
halofantrine and quinine are thought to have occurred by similar mechanisms.

Plasmodium have developed resistance against antifolate combination drugs, the most
commonly used being sulfadoxine and pyrimethamine. Two gene mutations are thought
to be responsible, allowing synergistic blockages of two enzymes involved in folate
synthesis. Regional variations of specific mutations give differing levels of resistance.

Atovaquone is recommended to be used only in combination with another anti-malarial


compound as the selection of resistant parasites occurs very quickly when used in mono-

24

PDF created with pdfFactory Pro trial version www.pdffactory.com


therapy. Resistance is thought to originate from a single-point mutation in the gene
coding for cytochrome-b.

Spread of resistance
There is no single factor that confers the greatest degree of influence on the spread of
drug resistance, but a number of plausible causes associated with an increase have been
acknowledged. These include aspects of economics, human behaviour, pharmokinetics,
and the biology of vectors and parasites.

The most influential causes are examined below:

1. The biological influences are based on the parasites ability to survive the presence
of an anti-malarial thus enabling the persistence of resistance and the potential for
further transmission despite treatment. In normal circumstances any parasites that
persist after treatment are destroyed by the host's immune system, therefore any
factors that act to reduce the elimination of parasites could facilitate the
development of resistance. This attempts to explain the poorer response
associated with immunocompromised individuals, pregnant women and young
children.
2. There has been evidence to suggest that certain parasite-vector combinations can
alternatively enhance or inhibit the transmission of resistant parasites, causing
'pocket-like' areas of resistance.
3. The use of anti-malarials developed from similar basic chemical compounds can
increase the rate of resistance development, for example cross-resistance to
chloroquine and amiodiaquine, two 4-aminoquinolones and mefloquine
conferring resistance to quinine and halofantrine. This phenomenon may reduce
the usefulness of newly developed therapies prior to large-scale usage.
4. The resistance to anti-malarials may be increased by a process found in some
species of plasmodium, where a degree of phenotypic plasticity was exhibited,
allowing the rapid development of resistance to a new drug, even if the drug has
not been previously experienced.
5. The pharmokinetics of the chosen anti-malarial are key; the decision of choosing
a long-half life over a drug that is metabolised quickly is complex and still
remains unclear. Drugs with shorter half-life's require more frequent
administration to maintain the correct plasma concentrations, therefore potentially
presenting more problems if levels of adherence and compliance are unreliable,
but longer-lasting drugs can increase the development of resistance due to
prolonged periods of low drug concentration.
6. The pharmokinetics of anti-malarials is important when using combination
therapy. Mismatched drug combinations, for example having an 'unprotected'
period where one drug dominates can seriously increase the likelihood of
selection for resistant parasites.
7. Ecologically there is a linkage between the level of transmission and the
development of resistance, however at present this still remains unclear.

25

PDF created with pdfFactory Pro trial version www.pdffactory.com


8. The treatment regime prescribed can have a substantial influence on the
development of resistance. This can involve the drug intake, combination and
interactions as well as the drug's pharmokinetic and dynamic properties.

Prevention of resistance
The prevention of anti-malarial drug resistance is of enormous public health importance.
It can be assumed that no therapy currently under development or to be developed in the
foreseeable future will be totally protective against malaria. In accordance with this, there
is the possibility of resistance developing to any given therapy that is developed. This is a
serious concern, as the rate at which new drugs are produced by no means matches the
rate of the development of resistance. In addition, the most newly developed therapeutics
tend to be the most expensive and are required in the largest quantities by some of the
poorest areas of the world. Therefore it is apparent that the degree to which malaria can
be controlled depends on the careful use of the current drugs to limit, insofar as it is
possible, any further development of resistance.

Provisions essential to this process include the delivery of fast primary care where staff
are well trained and supported with the necessary supplies for efficient treatment. This in
itself is inadequate in large areas where malaria is endemic thus presenting an initial
problem. One method proposed that aims to avoid the fundamental lack in certain
countries health care infrastructure is the privatisation of some areas, thus enabling drugs
to be purchased on the open market from sources that are not officially related to the
health care industry. Although this is now gaining some support there are many problems
related to limited access and improper drug use, which could potentially increase the rate
of resistance development to an even greater extent.

There are two general approaches to preventing the spread of resistance: preventing
malaria infections and, preventing the transmission of resistant parasites.

Preventing malaria infections developing has a substantial effect on the potential rate of
development of resistance, by directly reducing the number of cases of malaria thus
decreasing the requirement for anti-malarial therapy. Preventing the transmission of
resistant parasites limits the risk of resistant malarial infections becoming endemic and
can be controlled by a variety of non-medical methods including insecticide-treated bed
nets, indoor residual spraying, environmental controls (such as swamp draining) and
personal protective methods such as using mosquito repellent. Chemoprophylaxis is also
important in the transmission of malaria infection and resistance in defined populations
(for example travellers).

A hope for future of anti-malarial therapy is the development of an effective malaria


vaccine. This could have enormous public health benefits, providing a cost-effective and
easily applicable approach to preventing not only the onset of malaria but the
transmission of gametocytes, thus reducing the risk of resistance developing. Anti-
malarial therapy could be also be diversified by combining a potentially effective vaccine

26

PDF created with pdfFactory Pro trial version www.pdffactory.com


with current chemotherapy, thereby reducing the chance of vaccine resistance
developing.

Combination therapy

The problem of the development of malaria resistance must be weighed against the
essential goal of anti-malarial care; that is to reduce morbidity and mortality. Thus a
balance must be reached that attempts to achieve both goals whilst not compromising
either too much by doing so. The most successful attempts so far have been in the
administration of combination therapy. This can be defined as, 'the simultaneous use of
two or more blood schizonticidal drugs with independent modes of action and different
biochemical targets in the parasite'. There is much evidence to support the use of
combination therapies, some of which has been discussed previously, however several
problems prevent the wide use in the areas where its use is most advisable. These include:
problems identifying the most suitable drug for different epidemiological situations, the
expense of combined therapy (it is over 10 times more expensive than traditional mono-
therapy), how soon the programmes should be introduced and problems linked with
policy implementation and issues of compliance.

The combinations of drugs currently prescribed can be divided into two categories: Non-
artemesinin and Quinine based combinations and, Artemesinin based combinations. It is
also important to distinguish fixed-dose combination therapies (in which two or more
drugs are co-formulated into a single tablet) from combinations achieved by taking two
separate antimalarials.

Non-Artemisinin based combinations

Components Description Dose

This fixed-dose combination has been used


for many years and has widespread 25 mg/kg of
Sulfadoxine-
resistance. It causes few adverse effects but is sulfadoxine and
Pyrimethamine
cheap and is available in a single dose, thus 1.25 mg/kg of
(SP) (Fansidar)
decreasing problems associated with pyrimethamine.
adherence and compliance.

This is another cost-effective combination,


which benefits from the drugs having similar Chloroquine 25 mg/kg
SP plus pharmacokinetic profiles but different over 3 days with a
Chloroquine biochemical parasitic targets. High levels of single dose of SP as
resistance to one or both components means it described above.
is not effective in most locations.

27

PDF created with pdfFactory Pro trial version www.pdffactory.com


This combination has been shown to produce
a faster rate of clinical recovery than SP and
Chloroquine, however there are certain
adverse reactions associated with use that
10 mg/kg of
have limited its distribution. It is thought to
SP plus Amodiaquine per day
have a longer therapeutic lifetime than other
Amodiaquine for 3 days with a single
combinations and may be a more cost-
standard dose of SP.
effective option to introduce in areas where
resistance is likely to develop. This is
unlikely to occur until more information
regarding its safety has been obtained.

This is a single dose pill and offered obvious


SP plus advantages over more complex regimes but it
Mefloquine has not been recommended for used since
(Fansimef) 1990 due to widespread resistance to the
components.

Despite the increasing levels of resistance to


Quinine this combination has proven to be
particularly efficacious. The longer half-life
of the Tetracycline component ensures a high
cure rate. Problems with this regime include
the relatively complicated drug regimen, Quinine 10 mg/kg
Tetracycline or where Quinine must be taken every 8 hours doses every 8 hours
Doxycycline plus for 7 days. Additionally, there are severe side and Tetracycline in
Quinine effects to both drugs (Cinchonism in Quinine) 4 mg/kg doses every 6
and Tetracyclines are contraindicated in hours for 7 days.
children and pregnant women. For these
reasons this combination is not recommended
as first-line therapy but can be used for non-
responders who remain able to take oral
medication.

Artemesinin-based combination therapies (ACTs)

Artemesinin has a very different mode of action than conventional anti-malarials (see
information above), this makes is particularly useful in the treatment of resistant
infections, however in order to prevent the development of resistance to this drug it is
only recommended in combination with another non-artemesinin based therapy. It
produces a very rapid reduction in the parasite biomass with an associated reduction in
clinical symptoms and is known to cause a reduction in the transmission of gametocytes

28

PDF created with pdfFactory Pro trial version www.pdffactory.com


thus decreasing the potential for the spread of resistant alleles. At present there is no
known resistance to Artemesinin (though some resistant strains may be emerging) and
very few reported side-effects to drug usage, however this data is limited.

Components Description Dose

This combination has been


thoroughly tested in randomised
controlled trials and has
demonstrated that it is well
tolerated with few side effects.
However, chloroquine resistance
Artesunate and Chloroquine means that it is frequently
ineffective(i n one study there was
less than 85% cure in areas where
chloroquine resistance was
known). It is therefore not
approved by the WHO for use in
combination therapy.

This combination has also been


tested and proved to be more
Dosage is as a fixed-
efficacious and similarly well
dose combination
tolerated to the Chloroquine
(ASAQ)
combination. The cure rate was
Artesunate and Amodiaquine recommended as
greater than 90%, potentially
(Coarsucam and ASAQ) 4 mg/kg of Artesunate
providing a viable alternative
and 10 mg/kg of
where levels of Chloroquine
Amodiaquine per day
resistance are high. The main
for 3 days.
disadvantage is a suggested link
with neutropenia.

This has been used as an


The standard dose
efficacious first-line treatment
required is 4 mg/kg
regimen in areas of Thailand for
per day of Artesunate
many years. Mefloquine is known
Artesunate and Mefloquine plus 25 mg/kg of
to cause vomiting in children and
(Artequin and ASMQ) Mefloquine as a split
induces some neuropsychiatric and
dose of 15 mg/kg on
cardiotoxic effects, interestingly
day 2 and 10 mg/kg
these adverse reactions seem to be
on day three.
reduced when the drug is
combined with Artesunate, it is

29

PDF created with pdfFactory Pro trial version www.pdffactory.com


suggested that this is due to a
delayed onset of action of
Mefloquine. This is not considered
a viable option to be introduced in
Africa due to the long half-life of
Mefloquine, which potentially
could exert a high selection
pressure on parasites.

This combination has been


extensively tested in 16 clinical
trials, proving effective in children
under 5 and has been shown to be
better tolerated than Artesunate
plus Mefloquine combinations.
There are no serious side effects
Artemether and Lumefantrine
documented but the drug is not
(Coartem Riamet, and
recommended in pregnant or
Lonart)
lactating women due to limited
safety testing in these groups. This
is the most viable option for
widespread use and is available in
fixed-dose formulas thus
increasing compliance and
adherence.

This is a well tolerated It is recommended in


combination but the overall level doses of 4 mg/kg of
Artesunate and
of efficacy still depends on the Artesunate per day for
Sulfadoxine/Pyrimethamine
level of resistance to Sulfadoxine 3 days and a single
(Ariplus)
and Pyrimethamine thus limiting is dose of 25 mg/kg of
usage. SP.

Other combinations

There are several anti-malarial combinations currently being developed that are hoped to
be highly efficacious, cost-effective, safe and well tolerated. These are to be newly
developed compounds and not derivatives of currently used drugs, thus decreasing the
likelihood of resistance.

30

PDF created with pdfFactory Pro trial version www.pdffactory.com


Components Description Dose

dihydroartemisinin- Has been studied mainly in China, Vietnam and other


piperaquine (Duo- countries in SEAsia. The drug has been shown to be
Cotecxin, Artekin) highly efficacious (greater than 90%).

Has been tested and demonstrated a clinical response


pyronaridine and
rate of 100% in one trial in Hainan (an area with high
artesunate (Pyramax)
levels of P. falciparum resistance to Pyronaridine).

Is the most tested drug currently under development


and could be introduced in African countries
Chlorproguanil-Dapsone
imminently. It is not recommended as a monotherapy
and Artesunate (CDA)
due to concerns of resistance developing thus
threatening the future use of related compounds.

Experimental drugs
In 1996, Geoff McFadden became aware of the work of the biologist Ian Wilson, who
had discovered that the plasmodia responsible for causing malaria retained parts of
chloroplasts, an organelle usually found in plants, complete with their own functioning
genomes. This led Professor McFadden to the realisation that herbicides may be useful
lead compounds for the development of drugs against malaria. These "apicoplasts" are
thought to have originated through the endosymbiosis of algae and play a crucial role in
fatty acid bio-synthesis in plasmodia. To date, 466 proteins have been found to be
produced by apicoplasts and these are now being looked at as possible targets for novel
anti-malarial drugs.

Coartem
Coartem (artemether 20 mg/lumefantrine 120 mg) is an artemisinin-based
combination therapy (ACT) indicated for the treatment of acute uncomplicated
plasmodium falciparum malaria, the most dangerous form of the disease. Coartem
is produced by the Swiss pharmaceutical company, Novartis.

Coartem is comprised of two key ingredients; artemether, which is a derivative of


artemisinin, and lumefantrine (or benflumetol) an antimalarial drug. The combination
of these two ingredients has led to one of the most successful malaria treatments of its
kind. Coartem is a highly effective and well-tolerated malaria treatment, providing cure

31

PDF created with pdfFactory Pro trial version www.pdffactory.com


rates of up to 97%, even in areas of multi-drug resistance. In 2001, Coartem became the
first fixed dose ACT to meet the World Health Organization's (WHO) pre-qualification
criteria for efficacy, safety and quality.

In 2002, artemether-lumefantrine tablets were added to the WHO's Essential Medicines


list, , an index of essential drugs which help guide the purchasing decisions of Member
States and UN agencies.

Access To Treatment

Coartem is provided without profit to developing countries using grants from the Global
Fund to Fight AIDS, Tuberculosis and Malaria, US President’s Malaria Initiative along
with other donors. These broad partnerships have provided millions of children and
adults with access to a high-quality treatment for malaria.

Novartis has lowered the price of Coartem by 50% since 2001, increasing access to
patients around the world. The first significant price reduction occurred in 2006, when
the price of Coartem decreased from an average of US $1.57 to US $1.00. In 2006, due to
an improved supply situation for the natural ingredient artemisinin, Novartis was able to
undertake the pharmaceutical industry’s most aggressive manufacturing scale-up of its
kind from 4 million treatments in 2004 to 62 million treatments in 2006. Novartis and its
partners invested heavily in expanding production capacity at their state-of-the-art
facilities in China, and Suffern, New York. This increase in production capacity ensured
that supplies of Coartem met demand which enabled Novartis to further decrease the
price of Coartem.

In April 2008, Novartis further reduced the public sector price of Coartem by
approximately 20%, to an average of US $0.80 (or US $0.37 for a child’s treatment
pack). This price reduction was made possible through production efficiency gains.

Innovation of Coartem Dispersible

In January 2009, Novartis and Medicines for Malaria Venture (MMV) launched Coartem
Dispersible, the first ACT developed specifically for children suffering from malaria.
Coartem Dispersible contains the same amount of artemether and lumefantrine as
Coartem. A phase III study published in The Lancet showed that Coartem Dispersible
provides a high cure rate of 97.8%, which is comparable to that of Coartem (98.5%).
Investigators also reported that it had a good safety profile.

According to the WHO, there were 247 million cases of malaria in 2006, causing about
880,000 deaths, mostly among African children. In Africa alone, a child dies every 30
seconds from malaria. Before Coartem Dispersible, many parents and healthcare workers
crushed bitter-tasting antimalarial tablets for their children to swallow. The sweet-tasting
Coartem Dispersible tablets disperse quickly in small amounts of water, easing
administration and ensuring effective dosing.

32

PDF created with pdfFactory Pro trial version www.pdffactory.com


Novartis and MMV provide malaria case management educational programs, which
include hands-on training for local healthcare workers, customized training manuals, and
user-friendly packaging to ensure that Coartem Dispersible is properly used and to
improve patient compliance. Like Coartem, Coartem Dispersible is provided to the public
sector without profit to benefit those people most in need in the developing world.

Artemisinin

Pharmacokinetic data

Routes Oral

Artemisinin is a drug used to treat multi-drug resistant strains of falciparum malaria. The
compound (a sesquiterpene lactone) is isolated from the plant Artemisia annua. Not all
plants of this species contain artemisinin. Apparently it is only produced when the plant
is subjected to certain conditions, most likely biotic or abiotic stress. It can be synthesized
from artemisinic acid The drug is derived from a herb used in Chinese traditional
medicine, though it is usually chemically modified and combined with other medications.

Use of the drug by itself as a monotherapy is explicitly discouraged by the World Health
Organization as there have been signs that malarial parasites are developing resistance to
the drug. Combination therapies that include artemisinin are the preferred treatment for
malaria and are both effective and well tolerated in patients. The drug is also being
studied as a treatment for cancer.

History

33

PDF created with pdfFactory Pro trial version www.pdffactory.com


Artemisia has been used by Chinese herbalists for more than a thousand years in the
treatment of many illnesses, such as skin diseases and malaria. The earliest record dates
back to 200 BC, in the "Fifty-two Prescriptions" unearthed from the Mawangdui Han
Dynasty Tombs. Its antimalarial application was first described in Zhouhou Beji Fang
("The Handbook of Prescriptions for Emergencies"), edited in the middle of the fourth
century by Ge Hong. In the 1960s a research program was set up by the Chinese army to
find an adequate treatment for malaria. In 1972, in the course of this research, Tu Youyou
discovered artemisinin in the leaves of Artemisia annua (annual wormwood). The drug is
named Qinghaosu in Chinese. It was one of many candidates then tested by Chinese
scientists from a list of nearly 200 traditional Chinese medicines for treating malaria. It
was the only one that was effective, but it was found that it cleared malaria parasites from
their bodies faster than any other drug in history. Artemisia annua is a common herb and
has been found in many parts of the world, including along the Potomac River, in
Washington, D.C.

It remained largely unknown to the rest of the world for about ten years, until results
were published in a Chinese medical journal. The report was met with skepticism at first,
because the Chinese had made unsubstantiated claims about having found treatments for
malaria before. In addition, the chemical structure of artemisinin, particularly the
peroxide, appeared to be too unstable to be a viable drug.

Artemisinin derivatives
Because artemisinin itself has physical properties such as poor bioavailability that limit
its effectiveness, semi-synthetic derivatives of artemisinin, including artemether and
artesunate, have been developed.

Chemically modified analogues

There are a number of derivatives and analogues within the artemisinin family:

• Artesunate (water-soluble: for oral, rectal, intramuscular, or intravenous use)


• Artemether (lipid-soluble: for oral, rectal or intramuscular use)
• Dihydroartemisinin
• Artelinic acid
• Artenimol
• Artemotil

There are also simplified analogs in preclinical research.

Purely synthetic analogues

To counter the present shortage in leaves of Artemisia annua, researchers have been
searching for a way to develop artemisinin artificially in the laboratory. A 2006 paper in
Nature presented a genetically engineered yeast that can synthesize a precursor called
artemisinic acid which can be chemically converted to artemisinin.

34

PDF created with pdfFactory Pro trial version www.pdffactory.com


Indications
Malaria

Artemisinins can be used alone, but this leads to a high rate of recrudescence (return of
parasites) and other drugs are required to clear the body of all parasites and prevent
recurrence. The World Health Organization is pressuring manufacturers to stop making
the uncompounded drug available to the medical community at large, saying it would be
a significant loss if the malaria parasite developed resistance to Artemisinin.

The World Health Organisation has recommended that a switch to artemisinin


combination therapies (ACT) be made in all countries where the malaria parasite has
developed resistance to chloroquine. Artemisinin and its derivatives are now standard
components of malaria treatment in China, Vietnam, and some other countries in Asia
and Africa, where it has been proven to be a safe and effective anti-malarial treatment.
Fixed-dose combinations are preferred as this guarantees that the partner drug is present
to eradicate the last parasites while the artemisinin component removes the majority at
the start of the treatment.

A large number of fixed-dose ACTs are now available containing an artemisinin


component and a partner drug which has a long half-life, such as mefloquine (ASMQ),
lumefantrine (Coartem), amodiaquine (ASAQ), piperaquine (Duo-Cotecxin) and
antifolates (Ariplus). Most are made to GMP standard. A separate issue concerns the
quality of some artemisinin-containing products being sold in Africa and South-East
Asia.

Artemisinins are not used for malaria prophylaxis (prevention) because of the extremely
short activity of the drug. To be effective, it would have to be administered multiple
times each day.

Cancer treatment

Artemisinin is undergoing early research and testing for the treatment of cancer,
primarily by researchers at the University of Washington. Artemisinin has a peroxide
lactone group in its structure. It is thought that when the peroxide comes into contact with
high iron concentrations (common in cancerous cells), the molecule becomes unstable
and releases reactive oxygen species. It has been shown to reduce angiogenesis and the
expression of vascular endothelial growth factor in some tissue cultures.

Resistance
A study published in 2008 by Noedl and colleagues in the New England Journal of
Medicine suggests a consensus among researchers that artemisinin is losing its potency in
Cambodia and increased efforts are required to prevent drug-resistant malaria from

35

PDF created with pdfFactory Pro trial version www.pdffactory.com


spreading across the globe.. These findings were subsequently supported by a detailed
study from Western Cambodia.

Adverse effects
Artemisinins are generally well tolerated at the doses used to treat malaria. The side
effects from the artemisinin class of medications are similar to the symptoms of malaria:
nausea, vomiting, anorexia, and dizziness. Mild blood abnormalities have also been
noted. The only serious adverse effect is an allergic reaction. The drugs that are used in
combination therapies can contribute to the adverse effects that are experienced by those
undergoing treatment. Adverse effects in patients with acute falciparum malaria treated
with artemisinin derivatives tend to be higher.

Mechanism of action
There is no consensus regarding the mechanism through which artemisinin derivatives
kill the parasites. Their site of action within the parasite also remains controversial.

At the chemical level, one theory states that when the parasite that causes malaria infects
a red blood cell, it consumes hemoglobin within its digestive vacuole, liberating free
heme, an iron-porphyrin complex. The iron reduces the peroxide bond in artemisinin
generating high-valent iron-oxo species, resulting in a cascade of reactions that produce
reactive oxygen radicals which damage the parasite leading to its death.

Numerous studies have investigated the type of damage that oxygen radicals may induce.
For example, Pandey et al. have observed inhibition of digestive vacuole cysteine
protease activity of malarial parasite by artemisinin. These observations were supported
by ex vivo experiments showing accumulation of hemoglobin in the parasites treated with
artemisinin and inhibition of hemozoin formation by malaria parasites. Electron
microscopic evidence linking artemisinin action to the parasite's digestive vacuole has
been obtained showing that the digestive vacuole membrane suffers damage soon after
parasites are exposed to artemisinin.

Artemisinins have been reported to inhibit PfATP6, the parasite's SERCA-type enzyme
(calcium transporter), expressed in Xenopus oocytes. In this isolated system, resistance to
artemisinin is reported to be conferred by a single mutation in PfATP6. A study from
French Guiana in field isolates of malaria parasites identified an unrelated mutation in
PfATP6 that was associated with resistance to artemether. However this series of studies
does not constitute convincing evidence that PfATP6 is a site of action of artemisinins, or
that mutations in PfATP6 cause reduced artemisinin susceptibility. Robust evidence in
this context can be obtained by a transfection study, and it is notable that data from such a
study were presented at the Molecular Approaches to Malaria Conference (Lorne,
Australia) in February, 2008 yet remain unpublished. There is no evidence to suggest a
role for PfATP6 in mediating the artemisinin resistance that appears to be emerging in
Cambodia.

36

PDF created with pdfFactory Pro trial version www.pdffactory.com


A 2005 study investigating the mode of action of artemisinin using a yeast model
demonstrated that the drug acts on the electron transport chain, generates local reactive
oxygen species, and causes the depolarization of the mitochondrial membrane.

Dosing
The WHO approved adult dose of co-artemether (artemether-lumefantrine) for malaria is
4 tablets at 0, 8, 24, 36, 48 and 60 hours (six doses).This has been proven to be superior
to regimens based on amodiaquine. Artemesinin is not soluble in water and therefore
Artemisia annua tea was postulated not to contain pharmacologically significant amounts
of artemesinin. However, this conclusion was rebuked by several experts who stated that
hot water (85 oC), and not boiling water, should be used to prepare the tea. Although
Artemisia tea is not recommended as a substitute for the ACT (artemisinin combination
therapies) more clinical studies on artemisia tea preparation have been suggested.

Artesunate

Artesunate

Routes oral, IV, IM

Artesunate (INN) is part of the artemisinin group of drugs that treat malaria. It is a semi-
synthetic derivative of artemisinin that is water-soluble and may therefore be given by
injection. It is sometimes abbreviated AS.

Uses
Artesunate is used primarily as treatment for malaria; but it has also been shown to be
>90% efficacious at reducing egg production in Schistosoma haematobium infection.

Dosing
Intravenous dose of IV artesunate:

37

PDF created with pdfFactory Pro trial version www.pdffactory.com


• 2.4 mg/kg loading dose over 5 minutes
• 1.2 mg/kg dose 12 hours later
• 1.2 mg/kg once daily after that

Artesunate must always be given with another antimalarial such as mefloquine or


amodiaquine so as to avoid the development of resistance. The combination of
artesunate/amodiaquine has been found to be of equivalence to co-artemether.

Synthesis
Artesunate is prepared from dihydroartemisinin (DHA) by reacting it with succinic acid
anhydride in basic medium. Pyridine as base/solvent, sodium bicarbonate in chloroform
and catalyst DMAP (N,N-dimethylaminopyridine) and triethylamine in 1,2-
dichloroethane have been used, with yields of up to 100%. A large scale process involves
treatment of DHA in dichloromethane with a mixture of pyridine, a catalytic amount of
DMAP and succinic anhydride. The dichloromethane mixture is stirred for 6–9 h to get
artesunate in quantitative yield. The product is further re-crystallized from
dichloromethane. alpha-Artesunate is exclusively formed (m.p 135–137˚C).

Drug resistance
Clinical evidence of drug resistance has appeared in Western Cambodia, where
artemesinin monotherapy is common. There are as yet no reports of resistance emerging
elsewhere

Quinine
Pharmacokinetic data

Bioavailability 76 to 88%

Protein ~70%
binding

Metabolism Hepatic (mostly CYP3A4 and CYP2C19-


mediated)

Half life ~18 hours

38

PDF created with pdfFactory Pro trial version www.pdffactory.com


Excretion Renal (20%)

Routes Oral, intravenous

Quinine is a natural white crystalline alkaloid having antipyretic (fever-reducing),


antimalarial, analgesic (painkilling), and anti-inflammatory properties and a bitter taste.
It is a stereoisomer of quinidine which, unlike quinine, is an anti-arrhythmic.

Though it has been synthesized in the lab, the bark of the cinchona tree is the only
natural source of quinine. The medicinal properties of the cinchona tree were originally
discovered by the Quechua Indians of Peru and Bolivia; later, the Jesuits were the first to
bring the cinchona to Europe.

Quinine was the first effective treatment for malaria caused by Plasmodium falciparum,
appearing in therapeutics in the 17th century. It remained the antimalarial drug of choice
until the 1940s, when other drugs replaced it. Since then, many effective antimalarials
have been introduced, although quinine is still used to treat the disease in certain critical
situations. Quinine is available with a prescription in the United States and over-the-
counter, in very small quantities, in tonic water. Quinine is also used to treat lupus,
nocturnal leg cramps and arthritis.

Chemical structure
Quinine contains two major fused-ring systems: the aromatic quinoline and the bicyclic
quinuclidine.

Mechanism of action against P. falciparum


As with other quinoline anti-malarial drugs, the action of quinine has not been fully
resolved. The most widely accepted hypothesis of quinine action is based on the well-
studied and closely related quinoline drug, chloroquine. This model involves the
inhibition of hemozoin biocrystallization, which facilitates the aggregation of cytotoxic
heme. Toxic free heme accumulates in the parasites, leading to their death.

History
Quinine is an effective muscle relaxant, long used by the Quechua Indians of Peru to halt
shivering due to low temperatures. The Peruvians would mix the ground bark of cinchona
trees with sweetened water to offset the bark's bitter taste, thus producing tonic water.

Quinine has been used in unextracted form by Europeans since at least the early 1600s.
Quinine was first used to treat malaria in Rome in 1631. During the 1600s, malaria was
endemic to the swamps and marshes surrounding the city of Rome. Malaria was

39

PDF created with pdfFactory Pro trial version www.pdffactory.com


responsible for the death of several popes, many cardinals and countless common Roman
citizens. Most of the priests trained in Rome had seen malaria victims and were familiar
with the shivering brought on by the febrile phase of the disease. The Jesuit brother
Agostino Salumbrino (1561-1642), an apothecary by training who lived in Lima,
observed the Quechua using the bark of the cinchona tree for that purpose. While its
effect in treating malaria (and hence malaria-induced shivering) was unrelated to its
effect in controlling shivering from rigors, it was still a successful medicine for malaria.
At the first opportunity, Salumbrino sent a small quantity to Rome to test as a malaria
treatment. In the years that followed, cinchona bark was known as Jesuit's bark and
became one of the most valuable commodities shipped from Peru to Europe.

Synthetic quinine

Cinchona trees remain the only economically practical source of quinine. However, under
wartime pressure, research towards its synthetic production was undertaken. A formal
chemical synthesis was accomplished in 1944 by American chemists R.B. Woodward
and W.E. Doering. Since then, several more efficient quinine total syntheses have been
achieved, but none of them can compete in economic terms with isolation of the alkaloid
from natural sources. The first synthetic organic dye, mauveine, was discovered by
William Henry Perkin in 1856 while he was attempting to synthesize quinine.

Dosing and indication


As of 2006, quinine is no longer recommended by the WHO as first line treatment for
malaria and should only be used when artemesinins are not available.

Quinine is a basic amine and is therefore always presented as a salt. Various preparations
that exist include the hydrochloride, dihydrochloride, sulfate, bisulfate and gluconate.
This makes quinine dosing complicated since each of the salts has a different weight.

The following amounts of each form are equal:

• quinine base 100 mg


• quinine bisulfate 169 mg
• quinine dihydrochloride 122 mg
• quinine hydrochloride 111 mg
• quinine sulfate (actually (quinine)2H2SO4·2H2O) 121 mg
• quinine gluconate 160 mg.

All quinine salts may be given orally or intravenously (IV); quinine gluconate may
also be given intramuscularly (IM) or rectally (PR). The main problem with the rectal
route is that the dose can be expelled before it is completely absorbed; this can be
corrected by giving a half dose again.

40

PDF created with pdfFactory Pro trial version www.pdffactory.com


The IV dose of quinine is 8 mg/kg of quinine base every eight hours; the IM dose is
12.8 mg/kg of quinine base twice daily; the PR dose is 20 mg/kg of quinine base twice
daily. Treatment should be given for seven days.

Route of administration: Quinine is given either orally, intramuscular or intravenously.

Formulation/Strength: Tablet. 300mg


Ampoule I.M.I. 120mg/2ml or 600mg/10ml
Dosage:

Adult Give I.M. quinine (600mg/10ml) every 8 hours. (Three times daily)

- Small adult. 450mg (7.5ml)


(Under 50kg)

- Large adult. 600mg/10ml) every 8 hours.


(Over 50kg)

When the patients improves change to quinine tablet (300mg base) for 3 more days.

- Small adult. (Under 50kg) 450mg (1½ tab.) t.d.s.


- Large adults. (Over 50kg) 600mg (2 tab.) t.d.s.

NB: If quinine is given I.V. it must be diluted in dextrose/saline and SLOWLY infuse over 4
hours.

Children:
__________________________________________
Weight (kg) Quinine I.M/ml Quinine tab.
b.d t.d.s

3 – 3.9 ½ ¼
4 – 5.9 1 ¼
6 – 9.9 2 ½
15 – 19.9 3 ½
20 – 24.9 4 1
25 – 29.9 5 2
30 – 39.9 6 2½

The preparations available in the UK are quinine sulfate (200 mg or 300 mg tablets) and
quinine hydrochloride (300 mg/ml for injection). Quinine is not licensed for IM or PR
use in the UK. The adult dose in the UK is 600 mg quinine dihydrochloride IV or 600 mg
quinine sulfate orally every eight hours. For nocturnal leg cramps, the dosage is 200–
300 mg at night.

41

PDF created with pdfFactory Pro trial version www.pdffactory.com


In the United States, quinine sulfate is commercially available in 324-mg tablets under
the brand name Qualaquin; the adult dose is two tablets every eight hours. There is no
injectable preparation of quinine licensed in the U.S.: quinidine is used instead.

Side-effects
It is usual for quinine in therapeutic doses to cause cinchonism; in rare cases, it may even
cause death (usually by pulmonary edema). The development of mild cinchonism is not a
reason for stopping or interrupting quinine therapy and the patient should be reassured.
Blood glucose levels and electrolyte concentrations must be monitored when quinine is
given by injection. The patient should ideally be in cardiac monitoring when the first
quinine injection is given (these precautions are often unavailable in developing countries
where malaria is endemic).

Cinchonism is much less common when quinine is given by mouth, but oral quinine is
not well tolerated (quinine is exceedingly bitter and many patients will vomit after
ingesting quinine tablets): Other drugs such as Fansidar (sulfadoxine (sulfonamide
antibiotic) with pyrimethamine) or Malarone (proguanil with atovaquone) are often used
when oral therapy is required. Quinine ethyl carbonate is tasteless and odourless, but is
only commercially available in Japan. Blood glucose, electrolyte and cardiac monitoring
are not necessary when quinine is given by mouth.

Quinine can cause paralysis if accidentally injected into a nerve. It is extremely toxic in
overdose, and the advice of a poisons specialist should be sought immediately.

Quinine in some cases can lead to constipation, erectile dysfunction and diarrhea.

The New York Times Magazine described a case, presenting with fever, hypotension,
and blood abnormalities mimicking septic shock.

Abortifacient

Despite popular belief, quinine is an ineffective abortifacient (in the US, quinine is listed
as Pregnancy category D. Pregnant women who take toxic doses of quinine will suffer
from renal failure before experiencing any kind of quinine-induced abortion. Indeed,
quinine is the only drug recommended by the WHO as firstline treatment for
uncomplicated malaria in pregnancy.

Disease interactions

Quinine can cause hemolysis in G6PD deficiency, but again this risk is small and the
physician should not hesitate to use quinine in patients with G6PD deficiency when there
is no alternative. Quinine can also cause drug-induced immune thrombocytopenic
purpura (ITP). Symptoms can be severe enough to require hospitalisation and platelet
transfusion, with several cases resulting in death.

42

PDF created with pdfFactory Pro trial version www.pdffactory.com


Quinine can cause abnormal heart rhythms and should be avoided if possible in patients
with atrial fibrillation, conduction defects or heart block.

Quinine can worsen hemoglobinuria, myasthenia gravis and optic neuritis.

Hearing impairment

Some studies have related the use of quinine and hearing impairment, in particular
high-frequency loss, but it has not been conclusively established whether such
impairment is temporary or permanent.

Dihydroartemisinin (Second line drug)


Dihydroartemisinin (or dihydroqinghaosu) is a drug used to treat malaria.
Dihydroartemisinin is the active metabolite of all artemisinin compounds (artemisinin,
artesunate, artemether, etc.) and is also available as a drug in itself. The lactone of
artemisinin could selectively be reduced with mild hydride-reducing agents, such as
sodium borohydride, potassium borohydride, and lithium borohydride to
dihydroartemisinin (a lactol) in over 90% yield. It is a novel reduction, because normally
lactone cannot be reduced with sodium borohydride under the same reaction conditions
(0-5˚C, in methanol). Reduction with LiAlH4 leads to some rearranged products. It was
surprising to find that the lactone was reduced, but that the peroxy group survived.
However, the lactone of deoxyartemisinin resisted reduction with sodium borohydride
and could only be reduced with isobutylaluminium hydride to the lactol,
(deoxydihydroartimisinin). These results show that the peroxy group assists the reduction
of lactone with sodium borohydride to a lactol, but not to the alcohol which is the over-
reduction product. No clear evidence for this reduction process exists.

Dosing
Dihydroartemisinin is available as a fixed drug combination with piperaquine (each tablet
contains 40 mg of dihydroartemisinin and 320 mg of piperaquine; trade name Artekin,
manufactured by Holleykin Pharmaceuticals).

The adult dose is 1.6/12.8 mg/kg per dose (rounded up or down to the nearest half tablet)
given at 0 h, 8 h, 24 h, and 48 h. Alternatively, the same total dose may be given once
daily for three days.

Dihydroartemisinin is also sold in Africa as Cotecxin in 60 mg tablets, which is


manufactured by Zhejiang Holley Nanhu Pharmaceutical Co., Ltd., in China.

Piperaquine: (Second line drug)


Affiliation(s) du ou des auteurs / Author(s) Affiliation(s)

43

PDF created with pdfFactory Pro trial version www.pdffactory.com


Medicine Unit Fremantle and Pharmacology Unit Nedlands, School of Medicine and
Pharmacology, University of Western Australia, Crawley, Western Australia,
AUSTRALIE
Résumé / Abstract

Piperaquine is a bisquinoline antimalarial drug that was first synthesised in the 1960s,
and used extensively in China and Indochina as prophylaxis and treatment during the
next 20 years. A number of Chinese research groups documented that it was at least as
effective as, and better tolerated than, chloroquine against falciparum and vivax malaria,
but no pharmacokinetic characterisation was undertaken. With the development of
piperaquine-resistant strains of Plasmodium falciparum and the emergence of the
artemisinin derivatives, its use declined during the 1980s. However, during the next
decade, piperaquine was rediscovered by Chinese scientists as one of a number of
compounds suitable for combination with an artemisinin derivative. The rationale for
such artemisinin combination therapies (ACTs) was to provide an inexpensive, short-
course treatment regimen with a high cure rate and good tolerability that would reduce
transmission and protect against the development of parasite resistance. This approach
has now been endorsed by the WHO. Piperaquine-based ACT began as China-Vietnam 4
(CV4®: dihydroartemisinin [DHA], trimethoprim, piperaquine phosphate and
primaquine phosphate), which was followed by CV8® (the same components as CV4 but
in increased quantities), Artecom® (in which primaquine was omitted) and Artekin® or
Duo-Cotecxin® (DHA and piperaquine phosphate only). Recent Indochinese studies
have confirmed the excellent clinical efficacy of piperaquine-DHA combinations (28-day
cure rates >95%), and have demonstrated that currently recommended regimens are not
associated with significant cardiotoxicity or other adverse effects. The pharmacokinetic
properties of piperaquine have also been characterised recently, revealing that it is a
highly lipid-soluble drug with a large volume of distribution at steady
state/bioavailability, long elimination half-life and a clearance that is markedly higher in
children than in adults. The tolerability, efficacy, pharmacokinetic profile and low cost of
piperaquine make it a promising partner drug for use as part of an ACT.

Artemisia annua (Plant)


Artemisia annua

44

PDF created with pdfFactory Pro trial version www.pdffactory.com


Artemisia annua, also known as Sweet Wormwood, Sweet Annie, Sweet Sagewort or
Annual Wormwood (Chinese: pinyin: qīnghāo), is a common type of wormwood that is
native to temperate Asia, but naturalized throughout the world.

Medicinal uses
Sweet Wormwood was used by Chinese herbalists in ancient times to treat fever, but had
fallen out of common use, but was rediscovered in 1970 when the Chinese Handbook of
Prescriptions for Emergency Treatments (340 AD) was found. This pharmacopeia
contained recipes for a tea from dried leaves, prescribed for fevers (not specifically
malaria).

Extractions

In 1971, scientists demonstrated that the plant extracts had antimalarial activity in
primate models, and in 1972 the active ingredient, artemisinin (formerly referred to as
arteannuin), was isolated and its chemical structure described. Artemisinin may be
extracted using a low boiling point solvent such as diethylether and is found in the
glandular trichomes of the leaves, stems, and inflorescences, and it is concentrated in the
upper portions of plant within new growth.

Parasite treatment

It is commonly used in tropical nations which can afford it, preferentially as part of a
combination-cocktail with other antimalarials in order to prevent the development of
parasite resistance.

Malaria treatment

45

PDF created with pdfFactory Pro trial version www.pdffactory.com


Artemisinin itself is a sesquiterpene lactone with an endoperoxide bridge and has been
produced semi-synthetically as an antimalarial drug. The efficacy of tea made from
A. annua in the treatment of malaria is contentious. According to some authors,
artemesinin is not soluble in water and the concentrations in these infusions are
considered insufficient to treatment malaria. Other researchers have claimed that
Artemisia annua contains a cocktail of anti-malarial substances, and insist that clinical
trials be conducted to demonstrate scientifically that artemisia tea is effective in treating
malaria. This simpler use may be a cheaper alternative to commercial pharmaceuticals,
and may enable health dispensaries in the tropics to be more self-reliant in their malaria
treatment. In In 2004, the Ethiopian Ministry of Health changed Ethiopia’s first line anti-
malaria drug from Fansidar, a Sulfadoxine agent which has an average 36% treatment
failure rate, to CoArtem, an agent created from A. annua and which is 100% effective
when used correctly, despite a worldwide shortage at the time of the needed derivative
from A. annua.

Cancer treatment

The plant has also been shown to have anti-cancer properties. It is said to have the ability
to be selectively toxic to some breast cancer cells [Cancer Research 65:(23).Dec 1, 2005]
and some form of prostate cancer, there have been exciting preclinical results against
leukemia, and other cancer cells.

Mechanism

The proposed mechanism of action of artemisinin involves cleavage of endoperoxide


bridges by iron producing free radicals (hypervalent iron-oxo species, epoxides,
aldehydes, and dicarbonyl compounds) which damage biological macromolecules
causing oxidative stress in the cells of the parasite. Malaria is caused by the
Apicomplexan, Plasmodium falciparum, which largely resides in red blood cells and
itself contains iron-rich heme-groups (in the from of hemozoin).

Other uses
In modern-day central China, specifically Hubei Province, the stems of this wormwood
are used as food in a salad-like form. The final product, literally termed "cold-mixed
wormwood" is a slightly bitter salad with strong acid overtones from the spiced rice
vinegar used as a marinade. It is considered a delicacy and is typically more expensive to
buy than meat.

46

PDF created with pdfFactory Pro trial version www.pdffactory.com


Life Cycle of the Malaria Parasite

Credit: NIAID

Glossary

Diploid: Cells containing a full set


of chromosomes.
Gametes: Reproductive elements,
male and female.
Gametocytes: Precursors of the
sexual forms of the malaria

47

PDF created with pdfFactory Pro trial version www.pdffactory.com


Life Cycle of the Malaria or female gametes within the
Parasite stomach of the mosquito.
Haploid: Cells containing a half set
of chromosomes.
1. A female Anopheles mosquito carrying
Merozoite: The form of the
malaria-causing parasites feeds on a
malaria parasite that invades red
human and injects the parasites in the
blood cells.
form of sporozoites into the bloodstream.
Oocyst: A stage of the malaria
The sporozoites travel to the liver and
parasite within the mosquito which
invade liver cells.
is produced when male and female
2. Over 5-16 days*, the sporozoites grow,
gametes combine.
divide, and produce tens of thousands of
Ookinete: The actively moving
haploid forms, called merozoites, per liver
zygote of the malarial organism
cell. Some malaria parasite species remain
that penetrates the mosquito
dormant for extended periods in the liver,
stomach to form an oocyst under
causing relapses weeks or months later.
the outer gut lining.
3. The merozoites exit the liver cells and re-
Sporozoite: The infectious form of
enter the bloodstream, beginning a cycle
the malaria parasite, which is
of invasion of red blood cells, asexual
injected into people by mosquitoes.
replication, and release of newly formed
Zygote: The diploid cell resulting
merozoites from the red blood cells
from union of a male and a female
repeatedly over 1-3 days*. This
gamete.
multiplication can result in thousands of
parasite-infected cells in the host
bloodstream, leading to illness and complications of malaria that can last for
months if not treated.
4. Some of the merozoite-infected blood cells leave the cycle of asexual
multiplication. Instead of replicating, the merozoites in these cells develop into
sexual forms of the parasite, called male and female gametocytes, that circulate in
the bloodstream.
5. When a mosquito bites an infected human, it ingests the gametocytes. In the
mosquito gut, the infected human blood cells burst, releasing the gametocytes,
which develop further into mature sex cells called gametes. Male and female
gametes fuse to form diploid zygotes, which develop into actively moving
ookinetes that burrow into the mosquito midgut wall and form oocysts.
6. Growth and division of each oocyst produces thousands of active haploid forms
called sporozoites. After 8-15 days*, the oocyst bursts, releasing sporozoites into
the body cavity of the mosquito, from which they travel to and invade the
mosquito salivary glands. The cycle of human infection re-starts when the
mosquito takes a blood meal, injecting the sporozoites from its salivary glands
into the human bloodstream .

48

PDF created with pdfFactory Pro trial version www.pdffactory.com


FANSIDER
Fansidar is an antimalarial agent, each tablet containing 500 mg N1-(5,6-dimethoxy-4-
pyrimidinyl) sulfanilamide (sulfadoxine) and 25 mg 2,4-diamino-5-(p-chlorophenyl)-6-
ethylpyrimidine (pyrimethamine). Each tablet also contains cornstarch, gelatin, lactose,
magnesium stearate and talc.

INDICATION
Treatment of Acute Malaria
Fansidar is indicated for the treatment of acute, uncomplicated P. falciparum malaria for
those patients in whom chloroquine resistance is suspected. However, strains of P.
falciparum (see CLINICAL PHARMACOLOGY: Microbiology) may be encountered
which have developed resistance to Fansidar, in which case alternative treatment should
be administered.

Prevention of Malaria
Malaria prophylaxis with Fansidar is not routinely recommended and should only be
considered for travelers to areas where chloroquine-resistant P. falciparum malaria is
endemic and sensitive to Fansidar, and when alternative drugs are not available or are
contraindicated (see CONTRAINDICATIONS). However, strains of P. falciparum may
be encountered which have developed resistance to Fansidar.

DOSAGE AND ADMINISTRATION

The dosage should be swallowed whole, and not chewed, with plenty of fluids after a
meal.

Route of administration: The drug is given by oral route.

49

PDF created with pdfFactory Pro trial version www.pdffactory.com


Formulations/strength: Tablet. Fansider tablet containing sulphadoxine 500mg with
pyriethamine 25mg.

Dosage:

Children.

Weight (kg) Dose in Tablet

3–5 ¼
6–9 ½
10 – 19 1
20 – 29 1½
30 – 49 2 (adult)

- One single dose is given after quinine course.


- Used with quinine in the treatment of chloroquine resistant
- Do not use fansider by itself. Give it on day one with quinine.

Prevention of Malaria
The malaria risk must be carefully weighed against the risk of serious adverse drug
reactions (see INDICATIONS AND USAGE). If Fansidar is prescribed for prophylaxis,
it is important that the physician inquires about sulfonamide intolerance and points out
the risk and the need for immediate drug withdrawal if skin reactions do occur.

The first dose of Fansidar should be taken 1 or 2 days before arrival in an endemic area;
administration should be continued during the stay and for 4 to 6 weeks after return.

SIDE EFFECTAS

For completeness, all major reactions to sulfonamides and to pyrimethamine are included
below, even though they may not have been reported with Fansidar (see WARNINGS
and PRECAUTIONS: Information For The Patient).

Hematological Changes
Agranulocytosis, aplastic anemia, megaloblastic anemia, thrombocytopenia, leukopenia,
hemolytic anemia, purpura, hypoprothrombinemia, methemoglobinemia, and
eosinophilia.

Skin and Miscellaneous Sites Allergic Reactions


Erythema multiforme, Stevens-Johnson syndrome, generalized skin eruptions, toxic
epidermal necrolysis, urticaria, serum sickness, pruritus, exfoliative dermatitis,
anaphylactoid reactions, periorbital edema, conjunctival and scleral injection,
photosensitization, arthralgia, allergic myocarditis, slight hair loss, Lyell's syndrome,

50

PDF created with pdfFactory Pro trial version www.pdffactory.com


Gastrointestinal Reactions
Glossitis, stomatitis, nausea, emesis, abdominal pains, hepatitis, hepatocellular necrosis,
diarrhea, pancreatitis, feeling of fullness, and transient rise of liver enzymes.

Central Nervous System Reactions


Headache, peripheral neuritis, mental depression, convulsions, ataxia, hallucinations,
tinnitus, vertigo, insomnia, apathy, fatigue, muscle weakness, nervousness, and
polyneuritis.

Respiratory Reactions
Pulmonary infiltrates resembling eosinophilic or allergic alveolitis.

Genitourinary
Renal failure, interstitial nephritis, BUN and serum creatinine elevation, toxic nephrosis
with oliguria and anuria, and crystalluria.

Pregnancy
Teratogenic Effects: Pregnancy Category C
Fansidar has been shown to be teratogenic in rats when given in weekly doses
approximately 12 times the weekly human prophylactic dose. Teratology studies with
pyrimethamine plus sulfadoxine (1:20) in rats showed the minimum oral teratogenic dose
to be approximately 0.9 mg/kg pyrimethamine plus 18 mg/kg sulfadoxine. In rabbits, no
teratogenic effects were noted at oral doses as high as 20 mg/kg pyrimethamine plus 400
mg/kg sulfadoxine.

There are no adequate and well-controlled studies in pregnant women. However, due to
the teratogenic effect shown in animals and because pyrimethamine plus sulfadoxine may
interfere with folic acid metabolism, Fansidar therapy should be used during pregnancy
only if the potential benefit justifies the potential risk to the fetus.

Overdosage & Contraindications

WARNING
FATALITIES ASSOCIATED WITH THE ADMINISTRATION OF FANSIDAR HAVE OCCURRED DUE TO SEVERE
REACTIONS, INCLUDING STEVENS-JOHNSON SYNDROME AND TOXIC EPIDERMAL NECROLYSIS. FANSIDAR
PROPHYLAXIS MUST BE DISCONTINUED AT THE FIRST APPEARANCE OF SKIN RASH, IF A SIGNIFICANT
REDUCTION IN THE COUNT OF ANY FORMED BLOOD ELEMENTS IS NOTED, OR UPON THE OCCURRENCE OF
ACTIVE BACTERIAL OR FUNGAL INFECTIONS.

DRUG DESCRIPTION

51

PDF created with pdfFactory Pro trial version www.pdffactory.com


FATALITIES ASSOCIATED WITH THE ADMINISTRATION OF FANSIDAR
HAVE OCCURRED DUE TO SEVERE REACTIONS, INCLUDING STEVENS-
JOHNSON SYNDROME AND TOXIC EPIDERMAL NECROLYSIS. FANSIDAR
PROPHYLAXIS MUST BE DISCONTINUED AT THE FIRST APPEARANCE
OF SKIN RASH, IF A SIGNIFICANT REDUCTION IN THE COUNT OF ANY
FORMED BLOOD ELEMENTS IS NOTED, OR UPON THE OCCURRENCE OF
ACTIVE BACTERIAL OR FUNGAL INFECTIONS.

OVERDOSE
Acute intoxication may be manifested by headache, nausea, anorexia, vomiting and
central nervous system stimulation (including convulsions), followed by megaloblastic
anemia, leukopenia, thrombocytopenia, glossitis and crystalluria. In acute intoxication,
emesis and gastric lavage followed by purges may be of benefit. The patient should be
adequately hydrated to prevent renal damage. The renal, hepatic, and hematopoietic
systems should be monitored for at least 1 month after an overdosage. If the patient is
having convulsions, the use of parenteral diazepam or a barbiturate is indicated. For
depressed platelet or white blood cell counts, folinic acid (leucovorin) should be
administered in a dosage of 5 mg to 15 mg intramuscularly daily for 3 days or longer.

CONTRAINDICATIONS

• Repeated prophylactic (prolonged) use of Fansidar is contraindicated in patients


with renal or hepatic failure or with blood dyscrasias;
• Hypersensitivity to pyrimethamine, sulfonamides, or any other ingredient of
Fansidar;
• Patients with documented megaloblastic anemia due to folate deficiency;
• Infants less than 2 months of age;
• Prophylactic use of Fansidar in pregnancy at term and during the nursing period.

Mechanism of Action
Sulfadoxine and pyrimethamine, the constituents of Fansidar, are folic acid antagonists.
Sulfadoxine inhibits the activity of dihydropteroate synthase whereas pyrimethamine
inhibits dihydrofolate reductase.

52

PDF created with pdfFactory Pro trial version www.pdffactory.com


Activity in vitro
Sulfadoxine and pyrimethamine are active against the asexual erythrocytic stages of
Plasmodium falciparum. Fansidar may also be effective against strains of P. falciparum
resistant to chloroquine.

Pharmacokinetics
Absorption
After administration of 1 tablet, peak plasma levels for pyrimethamine (approximately
0.2 mg/L) and for sulfadoxine (approximately 60 mg/L) are reached after about 4 hours.

Distribution
The volume of distribution for sulfadoxine and pyrimethamine is 0.14 L/kg and 2.3 L/kg,
respectively.

Patients taking 1 tablet a week (recommended adult dose for malaria prophylaxis) can be
expected to have mean steady state plasma concentrations of about 0.15 mg/L for
pyrimethamine after about four weeks and about 98 mg/L for sulfadoxine after about
seven weeks. Plasma protein binding is about 90% for both pyrimethamine and
sulfadoxine. Both pyrimethamine and sulfadoxine cross the placental barrier and pass
into breast milk.

Metabolism
About 5% of sulfadoxine appears in the plasma as acetylated metabolite, about 2 to 3% as
the glucuronide. Pyrimethamine is transformed to several unidentified metabolites.

Elimination
A relatively long elimination half-life is characteristic of both components. The mean
values are about 100 hours for pyrimethamine and about 200 hours for sulfadoxine. Both
pyrimethamine and sulfadoxine are eliminated mainly via the kidneys.

SIDE EFFECTS: See also Warning section.


Nausea, vomiting, and loss of appetite may occur. This product may make you more
sensitive to the sun and sunburn (see also Precautions section). Less common side effects
may include headache, lightheadedness, trouble sleeping, tiredness, or irritability.

ARTEMISININ AND ITS DERIVATIVES


Artemisinin (qinghaosu) is the antimalarial principle isolated by Chinese scientists from
Artemisia annua L. It is a sesquiterpene lactone with a peroxide bridge linkage.
Artemisinin is poorly soluble in oils or water but the parent compound has yielded
dihydroartemisinin, the oil-soluble derivatives artemether and arteether, and the more
water-soluble derivatives sodium artesunate and artelinic acid. These derivatives have
more potent blood schizonticidal activity than the parent compound and are the most
rapidly effective antimalarial drugs known. They are used for the treatment of severe and

53

PDF created with pdfFactory Pro trial version www.pdffactory.com


uncomplicated malaria . They are not hypnozoiticidal but gametocytocidal activity has
been observed .

Formulations

A wide variety of formulations for oral or parenteral use or as suppositories are


available (see below). China and Viet Nam continue to be the main producers of
artemisinin and its derivatives.

Efficacy

The antimalarial activity of artemisinin and its derivatives is extremely rapid and most
patients show clinical improvement within 1-3 days after treatment. However, the
recrudescence rate is high when the drugs are used in monotherapy, depending on the
drug dose administered, the duration of treatment and the severity of disease, but not at
present on parasite resistance . Treatment for < 7 days gave unacceptably high
recrudescence rates . So far there is no confirmed in vivo evidence of resistance of P.
falciparum to artemisinin and its derivatives. The susceptibility of P. falciparum strains
from the China-Lao People’s Democratic Republic and China-Myanmar border areas to
various antimalarial drugs have been tested in vitro. The results have indicated declining
susceptibility of P. falciparum to artemisinin derivatives .

Under exceptional circumstances, such as when there is a history of an adverse reaction


to the combination agent, artemisinin monotherapy may be indicated, but a 7-day course
of therapy is recommended and efforts should be made to improve adherence to the
treatment. Preliminary results from Africa indicate that combinations of artesunate plus
amodiaquine or sulfadoxine-pyrimetha-mine are highly efficacious, although efficacy
may be compromised in areas with moderate to high levels of resistance to sulfadoxine-
pyrimethamine
P. Olliaro, personal communication)

These compounds are not recommended for use in the treatment of malaria due to P.
vivax, P. malariae or P. ovale since other effective antimalarial drugs are available for
this purpose. However, they may be used in the absence of micro-scopic diagnosis if they
are the recommended first-line treatment.

Use in pregnancy

Preclinical studies have consistently shown that artemisinin and its derivatives do not
exhibit mutagenic or teratogenic activity, but all of these drugs caused fetal resorption in
rodents at relatively low doses of 1/200-1/400 of the LD50,
i.e. > 10 mg/kg, when given after the sixth day of gestation . Reports on the use of these
drugs during pregnancy are limited . However, malaria can be particularly hazardous
during pregnancy. Artemisinin and its derivatives are therefore the drugs of choice for
severe malaria and can be used for treatment of uncomplicated malaria during the second
and third trimester of pregnancy in areas of multiple drug resistance . Owing to lack of

54

PDF created with pdfFactory Pro trial version www.pdffactory.com


data, their use in the first trimester is not recommended. The inadequacy of current
knowledge on the use of these drugs during pregnancy should be understood by
prescribers and all such use should, in principle, be monitored. Clinical outcomes of both
a successful and adverse nature should be reported to regulatory authorities.

Drug disposition

High-performance liquid chromatography-electron capture detection (HPLCECD) and


bioassay methods for studying the pharmacokinetics of artemisinin and its derivatives
have now been validated. HPLC-ECD detects separately the parent compound and the
major metabolite, dihydroartemisinin, whereas bioassays measure total activity, i.e.
parent compound plus metabolite(s). Both methods are cumbersome and only a limited
number of laboratories have the capability of conducting assays, especially using HPLC-
ECD, which requires a reductive-mode electrochemical analysis and must be performed
under oxygen-free conditions. An alternative HPLC method that uses ultraviolet
detection is somewhat easier and quicker to use. So far, all methods are for plasma only;
no method is available to measure levels in whole blood. With few exceptions, the lower
limit of detection of HPLC-based methods is = or <5mg/ml.

Oral bioavailability varies with the derivative and is influenced by disease status. All
derivatives, but not artemisinin itself are metabolized to a common bioactive metabolite,
dihydroartemisinin, at variable rates .

Adverse effects

Extensive clinical trials in China, Myanmar, Thailand and Viet Nam demonstrated no
acute cardiovascular or other vital organ toxicity. However, animal studies have
demonstrated severe neurotoxicity following parenteral administration of very high doses
of artemether or arteether. Both drugs produced a unique pattern of selective neuropathy
with chromatolysis and necrosis of scattered neurons in vestibular, motor and auditory
brain stem nuclei in rats, dogs and rhesus monkeys . Such effects have not been observed
with oral administration of any artemisinin derivative or with intravenous artesunate. This
has led to the suggestion that the effect is related to specific molecules and their route of
administration. The cause, however, appears to be due to sustainable high levels of the
drugs and their metabolites, which may occur following intramuscular injection, rather
than to the route of administration itself (T.G. Brewer, personal communication, 1996).

There is no clinical evidence to date of serious neurotoxicity resulting from the use of any
artemisinin drug in humans in prospective studies of more than 10 000 patients or in the
more than 2 million persons who have received these drugs . In Thailand, full
neurological examinations in more than 1 100 patients who had received an artemisinin
drug showed no specific pattern of neurological abnormalities. Studies in Thailand and
Viet Nam provided no evidence of any brain stem toxicity attributable to artemisinin and
artesunate . There is some concern about cerebellar dysfunction and prolonged or
repetitive treatment with artemisinin and its derivatives, which may occur in areas of high
transmission, must be viewed with caution. Additional studies to monitor subtle

55

PDF created with pdfFactory Pro trial version www.pdffactory.com


neurological changes and hearing loss are required, especially in patients undergoing
repetitive treatment. Post-marketing surveillance in countries where these drugs are
marketed and used is recommended.

A. ARTEMISININ

Formulations

• Tablets and capsules containing 250 mg of artemisinin (Viet Nam).


• Suppositories containing 100 mg, 200 mg, 300 mg, 400 mg or 500 mg of
artemisinin (Viet Nam).

Efficacy

Artemisinin is a sesquiterpene lactone with a peroxide bridge linkage that appears to be


responsible for its antimalarial activity. Artemisinin is a potent and rapidly acting blood
schizonticide, eliciting shorter parasite clearance times than chloroquine or quinine and
rapid symptomatic responses .

Artemisinin is poorly soluble in oils or water. Preclinical and clinical studies show that
artemisinin is effective against parasites resistant to all other operationally used
antimalarial drugs . It is not hypnozoiticidal. It reduces gametocyte carriage

Use

To reduce the recrudesence rate and the risk of development of resistance, as well as to
improve compliance, artemisinin should preferably be administered in combination with
another effective blood schizonticide. The use of artemisinin as monotherapy should be
limited to specific indications, such as in patients with a history of adverse reactions to
the combination drug. When mono-therapy is used, a 7-day course of therapy is
recommended and adherence to the treatment should be ensured.

When given as monotherapy to patients with uncomplicated falciparum malaria who have
some degree of immunity, a 5-day oral regimen of artemisinin has generally proven to be
curative.

Rectal formulations of artemisinin have a potentially important role to play in the


treatment of uncomplicated falciparum infections in children who vomit oral medication,
and as emergency treatment prior to referral in situations when parenteral antimalarial
drugs are not available or cannot be administered. Studies in Viet Nam have shown the
latter to be highly efficacious .

Recommended treatment

Although oral artemisinin has been widely employed in the treatment of uncomplicated
multidrug-resistant falciparum infections , very few well-designed dose-finding studies of

56

PDF created with pdfFactory Pro trial version www.pdffactory.com


artemisinin and its derivatives have been published. The dosage schedules indicated
below are based on available clinical data, as pharmacokinetic data are still insufficient
for formulating treatment regimens. When used as monotherapy, a minimum 7-day
course is required owing to the problem of recrudescent infections. If regimens of < 7
days are employed, combination with mefloquine is indicated to prevent such
recrudescence. Pharmacokinetic modelling suggests that a mefloquine dose of 25 mg/kg
provides better protection against development of resistance in combination therapy
regimens than one of 15 mg/kg (N. White, personal communication, 2000).

Monotherapy 20 mg/kg in a divided loading dose on the first day, followed by 10 mg/kg
once a day for 6 days.

Combination therapy

20 mg/kg in a divided loading dose on the first day, followed by 10 mg/kg once a day for
two more days plus mefloquine (15-25 mg of base per kg) as a single or split dose on the
second and/or third day.

In outpatient settings where adherence is questionable, combination with mefloquine (15


mg or 25 mg of base per kg) is indicated. Several clinical trials have shown that this is the
most effective treatment of multidrug-resistant P. falciparum malaria . Mefloquine is
administered on the second or third day because there is less risk of vomiting once the
clinical condition has improved.

Rectal administration

In emergency pre-referral treatment of severe malaria or for patients who cannot take oral
medication, artemisinin can be given by rectal administration before referral to hospital
or before medication becomes possible . This is intended as emergency management of
malaria in life-treatening circumstances and may be provided on a presumptive diagnosis
of malaria.

A single dose of 40 mg/kg should be given intarectally, then 20 mg/kg 24, 48 and 72
hours later, followed by oral treatment with an effective antimalarial drug.

Chemoprophylaxis

There is no rationale at present for using artemisinin for chemoprophylaxis.

Use in pregnancy

Artemisinin can be used for treatment of uncomplicated malaria during the second and
third trimester of pregnancy in areas of multidrug resistance . Owing to lack of data, use
in the first trimester of pregnancy is not recommended (see above).

Drug disposition

57

PDF created with pdfFactory Pro trial version www.pdffactory.com


Oral artemisinin is rapidly but incompletely absorbed with peak concentrations 1-2 h
after administration . Artemisinin is rapidly metabolized in vivo to dihydroartemisinin.
The elimination half-life is 2-5 hours . Bioavailability with rectal suppository
formulations is 30% less than with oral administration, although there is large inter-
individual variation. Studies comparing parasite clearance times following oral and rectal
administration have led to the conclusion that therapeutic concentrations should be
achieved with suppositories . Suppositories have been shown to be as effective as
parenteral anti-malarial drugs in clinical trials for the treatment of severe malaria .

Adverse effects

Adverse effects may include headache, nausea, vomiting, abdominal pain, itching, drug
fever abnormal bleeding and dark urine. Minor cardiac changes (mainly non-specific ST
changes and first degree atrioventricular block) have been noted during clinical trials.
These returned to normal after improvement of malaria symptoms. Experience indicates
that artemisinin and its derivatives are less toxic than the quinoline antimalarial drugs,
few adverse effects being associated with their use.

Prolonged or repetitive treatment with artemisinin and its derivatives must be treated with
caution. Additional studies, which monitor subtle neurological changes and hearing loss,
are required especially in patients undergoing repetitive treatment. Post-marketing
surveillance is recommended in countries where these drugs are marketed and used.

Contraindications

Artemisinin is not recommended in the first trimester of pregnancy because of limited


data.

Overdosage

There is no experience with overdosage with artemisinin.

B. ARTEMETHER

Formulations

• Capsules containing 40 mg of artemether (China).


• Composite tablets containing 50 mg of artemether (China).
• Ampoules of injectable solution for intramuscular injection containing 80 mg in

1 ml (China and France), or 40 mg in 1 ml for paediatric use (France).

Efficacy

58

PDF created with pdfFactory Pro trial version www.pdffactory.com


Artemether is an oil-soluble methyl ether derivative of dihydroartemisinin. As with
artemisinin, it is effective against P. falciparum resistant to all other operationally used
antimalarial drugs . It is not hypnozoiticidal but it reduces gametocyte carriage .

Use

As with artemisinin, when artemether is used for the treatment of uncomplicated P.


falciparum malaria, it should always be administered in combination with another
effective blood schizonticide to prevent recrudescence and delay the selection of resistant
strains. Monotherapy with oral or intramuscular artemether with a dose of 1-4 mg/kg per
day for 3-5 days results in an unacceptable rate of recrudescence . The use of artemether
as monotherapy should be limited to specific indications, such as in patients with a
history of adverse reactions to the combination drug. When monotherapy is used, a 7-day
course is recommended and efforts should be made to ensure adherence.

Artemether is not recommended for the treatment of malaria caused by P. vivax, P. ovale
and P. malariae since other effective antimalarial drugs are available for this purpose.
However, it may be used in the absence of microscopic diagnosis if the compound is the
recommended first-line treatment.

Recommended treatment

Because well-designed dose-finding studies of artemether are limited, the dosage


schedules outlined below for uncomplicated and severe malaria are based on available
clinical data. When used as monotherapy, a minimum 7-day course is required to prevent
recrudescence. If regimens of less than 7 days are employed, combination with
mefloquine or another effective blood schizonticide is indicated.

ARTEMETHER INJECTION:

Route of administration: Administered I.M. daily.

Formulation / Strength: 80mg

Dosage: - Day one 3.6mg/kg


- Day two – seven, 1.6mg/kg
- Give fansider on day three.

Weight (kg) Dosage ml (IMI) ml (IMI)


Day one Day 2-7

3 – 5.9 0.25 0.25


6 - 2.9 0.5 0.25
13 – 18.9 0.75 0.5
19 – 24.9 1ml 0.5
25 – 30.9 1.25 0.75
31 – 36.9 1.5 0.75

59

PDF created with pdfFactory Pro trial version www.pdffactory.com


37 – 43.9 1.75 1
Over 40 kg 2 1

• This drug is given IMI as a daily dose. You can change over to tablet once the
patient is able to take oral medication.
• Like the tablet, the first dose of injection is also double.

Uncomplicated malaria

Monotherapy: 4 mg/kg loading dose on the first day, followed by 2 mg/kg once a day for
6 days.

Combination therapy

4 mg/kg once a day for 3 days, plus mefloquine (15 mg or 25 mg of base per kg) as a
single dose or split dose on the second and/or third day.

Where adherence to the treatment is questionable, especially in outpatients, combination


with mefloquine is indicated . Cure rates of 95-98% have been demonstrated with this
combination in multidrug-resistant areas . Mefloquine is administered on the second or
third day because there is less risk of vomiting once the clinical condition has improved.

Severe malaria

3.2 mg/kg by the intramuscular route as a loading dose on the first day, followed by 1.6
mg/kg daily for a minimum of 3 days or until the patient can take oral therapy to
complete a 7-day course. The daily dose can be given as a single injection. In children,
the use of a tuberculin syringe is advisable since the injection volume will be small.

Use in pregnancy

Similar to artemisinin.

Drug disposition

The pharmacokinetics of artemether following oral administration appear to be similar to


those for artemisinin with mean peak plasma concentrations and mean plasma half lives
of 1-2 h and 2-3 h, respectively . The plasma concentrations of artemether are similar in
healthy subjects and those with acute uncomplicated malaria. Plasma antimalarial activity
is significantly greater with intramuscular administration than with oral use because the
first-pass biotransformation is bypassed . Bioavailability of artemether following
intramuscular administration was increased and clearance reduced in patients with acute
renal failure .

60

PDF created with pdfFactory Pro trial version www.pdffactory.com


Adverse effects

Toxicity studies in dogs and rats indicate that dose-dependent and potentially fatal
neurotoxic effects may occur after intramuscular injection of artemether at doses higher
than those used for malaria treatment . These changes can be widespread but mainly
affect areas associated with vestibular, motor and auditory functions . No similar findings
have been reported in humans treated with normal therapeutic doses of artemether.

Contraindications

Similar to artemisinin.

Overdosage

Similar to artemisinin.

C. ARTESUNATE

Formulations

• Tablets containing 50 mg of sodium artesunate (China, France and Viet Nam) or


200 mg of sodium artesunate (Switzerland).
• Ampoules for intramuscular or intravenous injection containing 60 mg of sodium
artesunate in 1 ml of injectable solution (China and Viet Nam).
• Suppositories of sodium artesunate (China).
• Rectal capsules containing 100 mg or 400 mg of sodium artesunate (Switzerland).

Efficacy

Artesunate, a water-soluble hemisuccinate derivative of dihydroartemisinin, is the most


widely used member of this family of drugs. It is unstable in neutral solutions and is
therefore only available for injections as artesunic acid. It is effective against P.
falciparum resistant to all other operationally used anti-malarial drugs . It does not have
hypnozoiticidal activity. It reduces gametocyte carriage rate .

Use

As with artemisinin, when artesunate is used for the treatment of uncomplicated

P. falciparum malaria, it should always be administered in combination with another


effective blood schizonticide to prevent recrudescence and delay the selection of resistant
strains. The use of artesunate as monotherapy should be limited to specific indications,
such as in patients with a history of adverse reactions to the combination drug. When
monotherapy is used, a 7-day course of therapy is recommended and efforts should be
made to ensure adherence.

61

PDF created with pdfFactory Pro trial version www.pdffactory.com


Artesunate is not recommended for the treatment of malaria caused by P. vivax,
P. ovale and P. malariae since other effective antimalarial drugs are available for this
purpose. However, it may be used in the absence of microscopic diagnosis if the
compound is the recommended first-line treatment.

Recommended treatment

Giving a dose twice daily offered no advantage over once daily dosing . While 7-day
regimens have a therapeutic advantage over 5-day regimens, this might be offset by
decreased patient adherence to the treatment; recrudescence rates of 50% are reported
following 3-day regimens regardless of the dosage used . The shorter courses provided
higher cure rates when a double dose was given on the first day of treatment or if the
drugs were combined with a longer-acting single-dose antimalarial such as mefloquine .
A regimen of 3-5 days of artesunate in combination with mefloquine given either
concomitantly or sequentially provides cure rates of nearly 100% .

ARTESUNATE TABLET

Dosage

Route of administration: Administer oral daily.

Dosage: Day one – 4mg/kg


Day two – seven – 2mg/kg
Give fansider on day three (3).

Weight (kg) Day.1.(tab.) Day.2-7 (tab.)

4 – 5.7 ½ ¼
6 – 8.9 ¾ ½
9 – 12.5 1 ½
12.6 – 18.5 1½ ¾
18.6 – 24.9 2 1
25 – 31.9 2½ 1½
32 – 37.5 3 1½
Over 37.5 4 2

Because well-designed dose-finding studies of artesunate are limited, dosage schedules


are based on available clinical data. When used as monotherapy, a minimum 7-day
course is required to prevent recrudescence. If regimens of less than 7 days are employed,
combination with mefloquine or another effective blood schizonticide is indicated. A
once-daily regimen has been shown to have similar parasite and fever clearance times as
a twice-daily regimen .

62

PDF created with pdfFactory Pro trial version www.pdffactory.com


Uncomplicated malaria

Monotherapy: 4 mg/kg loading dose on the first day, followed by 2 mg/kg once a day for
6 days.

Combination therapy

4 mg/kg once a day for 3 days, plus mefloquine (15 mg or 25 mg of base per kg) as a
single dose or split dose on the second and/or third day .

Where adherence to the treatment is questionable, especially in an outpatient situation,


combination with mefloquine (15 or 25 mg of base per kg) is indicated.

Severe malaria

2.4 mg/kg by the intramuscular route followed by 1.2 mg/kg at 12 and 24 h, then 1.2
mg/kg daily for 6 days. If the patient can swallow, the daily dose can be given orally.

2.4 mg/kg intravenously on the first day followed by 1.2 mg/kg daily until the patient can
take orally artesunate or another effective antimalarial drug.

Drug disposition

The pharmacokinetics of artesunate following oral administration appear to be similar to


those for artemisinin, with mean peak plasma concentrations and mean plasma half-lives
of 1-2 h and 2-3 h, respectively. The plasma concentrations of artesunate are more erratic
following administration by suppository compared to the intravenous route, but
inadequate absorption is unusual .

Adverse effects

Prospective clinical studies of more than 10 000 patients, and post-marketing surveillance
of over 4 600 patients in Thailand has not shown any serious drug-related adverse
reactions.

Rectal administration

In emergency pre-referral treatment of severe malaria or for patients who cannot take oral
medication, artesunate can be given by rectal administration before referral to hospital or
before oral medication becomes possible . This is intended as emergency pre-referral
management of malaria in life-threatening circumstances and may be provided to patients
on a presumptive diagnosis of malaria.

A single dose should be given rectally (rectal capsules/suppositories, 10 mg/kg) as soon


as possible once a diagnosis of malaria is made. If the rectal capsule is expelled within
the first hour, another rectal capsule should be inserted immediately. A second dose

63

PDF created with pdfFactory Pro trial version www.pdffactory.com


might be required 24 h after the first dose if the patient is still unable to take oral
medication at that time, and has not been able to access recommended parenteral
treatment.

There is no information on efficacy in patients with severe and complicated malaria who
have organ and systems failure, including renal failure and liver disease. No studies have
been undertaken with this formulation in pregnant or lactating women or in patients with
diarrhoea.

Rectal artesunate should not be given for the prevention of malaria.

D. DIHYDROARTEMISININ

Formulations

• Tablets containing 20 mg, 60 mg or 80 mg of dihydroartemisinin (China).


• Suppositories containing 80 mg of dihydroartemisinin (China).

Efficacy

Dihydroartemisinin is the active metabolite of artemisinin and its derivatives.

These derivatives have more potent blood schizonticidal activity than the parent
compound. Dihydroartemisinin is the most potent antimalarial of this group of
compounds but it is also the least stable.

Oral dihydroartemisinin has been shown to be effective in the treatment of multidrug-


resistant uncomplicated P. falciparum malaria in China, but experience outside that
country is limited . Recent studies in Thailand demonstrated a cure rate of 90% in 52
patients given 120 mg of dihydroartemisinin followed by 60 mg once daily for 7 days, i.e.
a total adult dose of 480 mg (S. Looareesuwan, personal communication, 1995).

Dihydroartemisinin does not have activity against hypnozoites. It reduces gametocyte


carriage rate.

Use

Dihydroartemisinin appears to offer no advantage over artesunate or artemether for the


treatment of uncomplicated or severe malaria.

Dihydroartemisinin is not recommended for the treatment of malaria caused by


P. vivax, P ovale and P. malariae since other effective antimalarial drugs are available
for this purpose. However, it may be used in the absence of micro-scopic diagnosis if the
compound is the recommended first-line treatment.

Recommended treatment

64

PDF created with pdfFactory Pro trial version www.pdffactory.com


4 mg/kg in a divided loading dose on the first day followed by 2 mg/kg daily for 6 days.

Data on dihydroartemisinin are very limited, but the currently recommended dosage is as
shown above. Dihydroartemisinin has been used in combination with mefloquine . Short
courses of treatment of less than 5 days have higher recrudescence rates .

Drug disposition

Oral dihydroartemisinin is rapidly absorbed and has a short elimination half-life although
little is known of its metabolism. Peak plasma concentrations are achieved in 1-2 h and
the drug disappears from the circulation within 8-10 h.

E. ARTEETHER

Formulations

Ampoules containing 150 mg of arteether in 2 ml of injectable solution (India,


Netherlands).

Efficacy

Arteether is the oil-soluble ethyl derivative of dihydroartemisinin. Clinical trials in India


have indicated that it is an effective and rapidly-acting drug for the treatment of
uncomplicated (275) and severe falciparum malaria .

Use

When arteether is used for the treatment of uncomplicated P. falciparum malaria, it


should always be administered in combination with another effective blood schizonticide
to improve its efficacy and delay the selection of resistant strains. A recrudescence rate of
6-14% has been observed with the use of alpha, beta-arteether . The use of arteether as
monotherapy should therefore be limited to specific indications, such as in patients with a
history of adverse reactions to the combination drug. When given as monotherapy, a 7-
day course is recommended and efforts should be made to ensure adherence.

Arteether is not recommended for the treatment of malaria caused by P. vivax, P. ovale
and P. malariae since other effective antimalarial drugs are available for this purpose.

Recommended treatment

The recommended regimen according to the Manufacturer is:

For adults, 150 mg/day administrated once daily by the intramuscular route for 3 days.

For children, 3 mg/kg per day by the intramuscular route for 3 days.

65

PDF created with pdfFactory Pro trial version www.pdffactory.com


Drug disposition

Intramuscular arteether has the lowest bioavailability (34%) of all the artemisinin
derivatives tested in the rat, with approximately 14% converted to dihydroartemisinin. It
has a long elimination half-life (> 20 h) and is more stable and more lipophilic than the
other artemisinin compounds.

Adverse effects

Animal studies have demonstrated limited symptomatic and pathological evidence of


neurotoxicity following parenteral administration of high doses (8-24 mg/kg per day for
14 days) of either arteether or artemether . Both drugs produced a unique pattern of
selective neuronopathy with chromatolysis and necrosis of scattered neurons in
vestibular, motor and auditory brain stem nuclei in rats, dogs and rhesus monkeys .

F. ARTELINIC ACID

Efficacy

Artelinic acid is a water-soluble derivative of artemisinin and is thought to be more stable


than artesunate in solution thus offering the potential for oral administration. The
compound is still under investigation. It is the only preparation undergoing transdermal
studies .

__________________________________________________________________

66

PDF created with pdfFactory Pro trial version www.pdffactory.com

You might also like