Você está na página 1de 36

AFRICAN SLEEPING SICKNESS Ibn Khaldoun, who wrote about the disease in his account of the

history of North Africa. The next report came from Guinea in 1734.
In 1803, the diseases that caused visible swollen lymph glands in
INTRODUCTION: The Disease and Importance West Africa came to be known as Winterbottom's sign, after the
description of the disease by Cris Winterbottom. Slave traders who
Human African trypanosomiasis (HAT), also called avoided trading and buying slaves who displayed those symptoms
sleeping sickness, is an illness endemic to sub-Saharan Africa. It is readily recognized such signs.
caused by the flagellate protozoan Trypanosoma brucei, which exists
in 2 morphologically identical subspecies: Trypanosoma brucei The earliest detection of trypanosomes in human blood was
rhodesiense (East African or Rhodesian African trypanosomiasis) and in 1902, when R.M. Forde discovered what was then thought to be
Trypanosoma brucei gambiense (West African or Gambian African filiaria in the blood of a steamboat captain who had traveled
trypanosomiasis). Both of these parasites are transmitted to human extensively along the River Gambia. J.H. Cook in East Africa made
hosts by bites of infected tsetse flies (Glossina palpalis transmits T similar discoveries of filiaria-like organisms in the blood, but
brucei gambiense and Glossina morsitans transmits T brucei confusion arose as to how filiaria worms could cause such varying
rhodesiense), which are found only in Africa. clinical symptoms. J.E. Dutton first correctly identified the parasite as
a trypanosome and subsequently named it Trypanosoma gambiense.
The reservoirs of infection for these vectors are exclusively In 1902, A. Castellani observed the presence of trypanosomes in
human in West African trypanosomiasis. However, East African cerebrospinal fluid taken from a sleeping sickness patient, but it
trypanosomiasis is a zoonotic infection with animal vectors. African wasn't until 1903 that D. Bruce correctly recognized that
trypanosomiasis is distinct from American trypanosomiasis, which is trypanosomes were the causative agents of sleeping sickness
caused by Trypanosoma cruzi and has different vectors, clinical transmitted to humans by tsetse flies, and that “trypanosome fever”
manifestations, and therapies. and “sleeping sickness” - both thought to be different diseases at the
time - were in fact the same.
The major epidemiology factor in African trypanosomiasis
is contact between humans and tsetse flies. An increasing tsetse fly Morphologically indistinguishable from the West African
density, changing feeding habits, expanding human development into species as well as the animal infecting species Trypanosoma brucei
tsetse fly–infested areas, and an increasing number of brucei, Trypanosoma brucei rhodensiense was first discovered in
immunologically naïve persons in previously endemic areas, Zambia by J.W.W. Stephens and H.B. Fantham in 1910. By 1926,
influences this interaction. Major outbreaks from 1920-1950 led to T.b. rhodensiense could be found along the fly-belt between Tabora
extensive treatment and, apparently, immunity for 50 years. Now, and Kigoma, Tanzania. The difficulties in identifying this virulent
infection is occurring again as the same populations lose their form of sleeping sickness lead to uncertainties today regarding the
immunity. evolution and progression of T.b. rhodensiense through the continent,
although it is generally agreed upon that it originated from the West
African form.
Trypanosomes are parasites with a 2-host life cycle:
mammalian and arthropod. The life cycle starts when the
trypanosomes are ingested during a blood meal by the tsetse fly from The earliest recorded major epidemics of sleeping sickness
a human reservoir in West African trypanosomiasis or an animal took place in Uganda and Congo between 1896 and 1908, where
reservoir in the East African form. The trypanosomes multiply over a roughly 500,000 people were estimated to have died in the Congo
period of 2-3 weeks in the fly midgut; then, the trypanosomes migrate Basin and approximately 300,000 died in Busoga, Uganda. With the
to the salivary gland, where they develop into epimastigotes. The Rift Valley transecting the country, Uganda is in the precarious
metacyclic trypomastigotes infect humans. position of having foci of both forms of diseases which resulted in
two other major epidemics of sleeping sickness - one in the late
1940's and another in 1980. Throughout West Africa, smaller
Although human sleeping sickness may not seem as epidemics of sleeping sickness rapidly spread from Senegal to
important on the world stage as diseases such as malaria and AIDS, it Cameroon during the 1920's, and died down by the late 1940's.
is nevertheless an important disease in Sub-Saharan Africa and is
responsible for a considerable degree of suffering and mortality in
countries where it is endemic. Some 55 million people in 37 countries ETIOLOGY
are at risk, with an estimated 50,000 new cases reported annually.
Left untreated, the outcome of the disease for the individual is death,
but equally insidious is the effect on communities and quality of life There are two clinical forms of African trypanosomiasis
resulting from the debilitating symptoms. In public health terms, the (also called sleeping sickness); each is named for the region of Africa
effects of the disease on the community life and, in particular, the in which they were found historically.
contribution of individuals to food production and community
support can be measured in terms of disability-adjusted life years lost East African sleeping sickness is caused by the parasite
(DALYS). Human sleeping sickness is responsible for 1.78 million Trypanosoma brucei rhodesiense, commonly found in Uganda,
DALYS, and the magnitude of this effect can be seen when compared Kenya, Tanzania, Malawi, Ethiopia, Zaire, Zimbabwe and Botswana.
with the related disease leishmaniasis, which has a value of 2.06
million DALYS despite there being 350 million people at risk in 88 East African sleeping sickness is an acute or rapidly
countries. progressing disease that typically leads to death within weeks or
months if not treated. The initial bite leaves a distinctive sore spot
Early History called a chancre. Symptoms, which appear one to four weeks after
infection, may include swollen lymph nodes, irritability, fever, severe
The earliest recorded account of sleeping sickness comes headache, fatigue, muscle and joint pain, and a skin rash. During the
from upper Niger during the 14th century in the historical writings of second stage of the disease, the parasite crosses the blood-brain
barrier and attacks the central nervous system. Neurological
1
complications include slurred speech, confusion, and difficulty with The surface of the trypanosome has numerous membrane-
walking. associated transport proteins for obtaining nucleic acid bases,
glucose, and other small molecular weight nutrients. None of these
West African Sleeping Sickness also called Gambian proteins reacts well with antibodies, because although they lie in
sleeping sickness is caused by a parasite called Trypanosoma brucei exposed regions of membrane, they are shielded by allosteric
gambiense commonly found in Democratic Republic of Congo, interference provided by the variant surface glycoprotein (VSG) coat
Angola, Sudan, Central African Republic, Republic of Congo, Chad proteins. This flagellated stage enters the bloodstream through the
and Northern Uganda. lymphatics and divides further, producing a patent parasitemia. The
number of parasites in the blood is generally so low that diagnosis by
microscopic examination is often negative. At some point,
Like East African sleeping sickness, West African trypanosomes enter the central nervous system, with serious
(Gambian) sleeping sickness is a serious slow developing disease that pathological consequences for humans. Some parasites transform into
is fatal if not treated. However, symptoms may not appear for months the non-dividing short, stumpy form, which has biochemistry similar
to years after the initial infection in a chronic disease. Initial to those of the long, slender form and the form found in the insect
symptoms include swollen lymph nodes, swelling of face and hands, vector.
skin rash, fever, severe headaches, muscle and joint pain, and fatigue.
Weight loss is common as the disease progresses. Neurological
impairment occurs during the second stage in which the victim can The tsetse fly becomes infected by ingesting a blood meal
experience personality changes, slurred speech, changes in sleep from an infected host. These short, stumpy forms are pre-adapted to
patterns, progressive confusion, difficulty with walking, and seizures. the vector, having a well-developed mitochondrion with a partial
TCA cycle. In the insect vector, the trypanosomes develop into
procyclic trypomastigotes in the midgut of the fly, and continue to
BACTERIOLOGY divide for approximately 10 days. Here they gain a fully functional
cytochrome system and TCA cycle. When the division cycles are
completed, the organisms migrate to the salivary glands, and
The Trypanosome transform into epimastigotes. These forms, in turn, divide and
transform further into metacyclic trypanosomes, the infective stage
Trypanosomes have been around for more than 300 million for humans and reservoir hosts. The cycle in the insect takes 25-50
years. They are microscopic spindle-shaped unicellular protozoan of days, depending upon the species of the fly, the strain of the
genus Trypanosoma, which are ubiquitous parasite of insects, plants, trypanosome, and the ambient temperature. If tsetse flies ingest more
birds, bats, fish, amphibians and mammals. Because they have been than one strain of trypanosome, there is the possibility of genetic
around for so long, they and their natural hosts have evolved together exchange between the two strains, generating an increase in genetic
to ensure their mutual survival. Trypanosomes can be found from diversity in an organism that may not have a sexual cycle.
Nairobi to New York, from Sydney to San Francisco, and from
Birmingham to Buenos Aires. Fortunately, few species of Flies can remain infected for life (2-3 months). Tsetse flies
trypanosomes are pathogenic. Trypanosomes, and other parasites, inject over 40,000 metacyclic trypanosomes when they take a blood
mainly cause disease when they spread to new hosts, like humans and meal. The minimum infective dose for most hosts is 300-500
their domestic animals, especially recent imports into endemic areas organisms, although experimental animals have been infected with a
of species that diverged since continent separated. single organism. Infection can also be acquired by eating raw meat
from an infected animal. In East Africa, this mode of transmission
African trypanosomes are extracellular organisms, both in may be important in maintaining the cycle in some reservoir hosts.
the mammalian and insect host. T. b. gambiense and T. b.
rhodesiense are morphologically indistinguishable, measuring 25-40 We generally associate trypanosomes with disease in
µ m in length. Infection in the human host begins when the infective Africa and South America. African trypanosomiasis is commonly
stage, known as the metacyclic stage, is injected intradermally by the known as Sleeping Sickness in humans and Nagana (meaning “loss
tsetse fly. The organisms rapidly transform into blood-stage of spirit” in Zulu language) in cattle.
trypomastigotes (long, slender forms), and divide by binary fission in
the interstitial spaces at the site of the bite wound. The buildup of
metabolic wastes and cell debris leads to the formation of a chancre. Many trypanosomes do not appear to harm their hosts, but
a number of species cause serious disease in humans or domestic
animals. Trypanosoma brucei gambiense and Trypanosoma brucei
Trypanosomes have a single specialized mitochondrion rhodesiense causes African sleeping sickness and is transmitted
called a kinetoplast mitochondrion. One of its unusual features is
that the entire DNA of the mitochondrion, which can be up to 25% of
the total cell DNA, is localized in the kinetoplast, adjacent to the Trypanosomes in the blood of vertebrates have been
flagellar pocket. Kinetoplast DNA or kDNA exists in two forms: observed to have three body types; a short, broad form often without
mini-circles and maxi-circles. Mini-circle DNA encodes guide RNAs a flagellum, called the promastigote; a long and narrow form, the
that direct extensive editing of RNA transcripts post-transcriptionally. trypomastigote, and an intermediate between these two, the
Maxi-circle DNA contains sequences that, when edited, direct epimastigote. In arthropod hosts, all developmental stages have been
translation of typically mitochondrially encoded proteins. reported to derive from an amastigote stage, which lacks a flagellum
and has a circular shape. Trypomastigote form may be active and
proliferative, at this stage they are referred to as metacyclic, which is
In the vertebrate host, trypanosomes depend entirely upon presumed to be the infective stage for vertebrates.
glucose for energy and are highly aerobic, despite the fact that the
kinetoplast-mitochondrion completely lacks cytochromes. Instead,
mitochondrial oxygen consumption is based on an alternative oxidase • Amastigote - Basal body anterior of nucleus, with a short,
that does not produce ATP. When in the insect vector, the parasite essentially non-functional, flagellum.
develops a conventional cytochrome chain and TCA cycle.

2
• Promastigote - Basal body anterior of nucleus, with a long • T. brucei gambiense - Causes slow onset chronic
detached flagellum. trypanosomiasis in humans. Most common in central and
• Epimastigote - Basal body anterior of nucleus, with a long western Africa, where humans are thought to be the
primary reservoir.
flagellum attached along the cell body.
• T. brucei rhodesiense - Causes fast onset acute
• Trypomastigote - Basal body posterior of nucleus, with a
trypanosomiasis in humans. Most common in southern and
long flagellum attached along the cell body.
eastern Africa, where game animals and livestock are
thought to be the primary reservoir.
These names are derived from the Greek “mastig”- • T. brucei brucei - Causes animal African trypanosomiasis,
meaning whip, referring to the trypanosome's whip-like flagellum. along with several other species of trypanosoma. T. b.
brucei is not human infective due to its susceptibility to
T. brucei is found as a trypomastigote in the slender, lysis by human apolipoprotein L1. However, as it shares
stumpy, procyclic and metacyclic forms. The procylic form many features with T. b. gambiense and T. b. rhodesiense
differentiates to the proliferitive epimastigote form in the salivary (such as antigenic variation) it is used as a model for
glands of the insect. human infections in laboratory and animal studies.

Subs-species of Trypanosome brucei These obligate parasites have two hosts - an insect vector
and mammalian host. Because of the large difference between these
Trypanosoma brucei spp. is an extracellular gram-negative hosts, the trypanosome undergoes complex changes during its life
parasitic protist species that causes African trypanosomiasis (or cycle to facilitate its survival in the insect gut and the mammalian
sleeping sickness) in humans and nagana in animals in Africa. There bloodstream. It also features a unique and notable variable surface
are 3 sub-species of T. brucei; T. b. brucei, T. b. gambiense and T. b. glycoprotein (VSG) coat in order to avoid the host's immune system.
rhodesiense. There is an urgent need for the development of new drug therapies, as
current treatments can prove fatal to the patient as well as the
trypanosomes

MORPHOLOGY

A sound knowledge of the basic features of the various trypanosomes enables the identification of each species and so the exact cause of
the disease. Once the basic features possessed by all trypanosomes are appreciated, the diagnostic differences can be recognized and the species
identified.

Basic morphology of trypanosomes

Diagram of a trypanosome Fig. 1

The parasite consists of a single cell varying in size from 8 take place within this unicellular organism — nutrition, respiration,
to over 50 μm. All the activities associated with a living organism excretion, reproduction. The substance of which all living cells

3
consist, the protoplasm, comprises three parts, an outer protective and For specific identification, a number of trypanosomes
retaining layer, the pellicle = cell envelope = cell membrane, within should be examined systematically for the presence or absence, size
which the cytoplasm forms the bulk of the contents. Suspended in the and position of a number of features:
cytoplasm are various structures, the most prominent being the
nucleus, which may be regarded as the command centre of the cell
and which also plays a major part in reproduction. It contains DNA • Presence or absence of trypanosomes of different
(deoxyribonucleic acid), which is arranged in the form of genes and appearance. If all individual trypanosomes are alike, the
chromosomes; it represents the genetic information and is responsible infection is called monomorphic (of one form); if there are
for the manufacture of enzymes and other proteins of the cell. distinctly different types it can be either a polymorphic (=
pleiomorphic) species, or a mixed infection of different
species.
Small granules (formerly called “volutin granules”) can
sometimes be seen in the cytoplasm; they may have various origins, • Presence or absence of a free flagellum. In certain species,
they may be food or nuclear reserves, or result from a reaction there may be some trypanosomes with, and some without, a
between the trypanosome and the host's immune system. free flagellum.
• Size of the trypanosome (expressed in μm).
Trypanosomes are thoroughly adapted to living and moving • The size and position of the kinetoplast. The position is
in the blood plasma or tissue fluid of the host. They are elongated and related to proximity to the posterior extremity (rear end) of
streamlined, and tapered at both ends. The pellicle, the outer layer of the organism.
the cytoplasm, is flexible enough to permit a degree of body • The degree of development of the undulating membrane. It
movement, while retaining a definite shape. As shown in Figure 1 a may be conspicuous or inconspicuous.
flagellum arises near to the posterior end from a parabasal body, and
runs the length of the trypanosome; it may be continued beyond the
• The shape of the parasite, particularly the shape of its
posterior part. The posterior extremity may vary from blunt
anterior end of the body as a whip-like free flagellum. Along the
to pointed.
length of the body, the pellicle and cytoplasm are pinched up into a
thin sheet of tissue called the undulating membrane, through the outer
margin of which runs the flagellum, as shown in Figure 1. Specific morphology

Among other basic morphological features, a distinct well- The sub genus Trypanozoon (the brucei group). This group
defined body, the kinetoplast, is seen near to the posterior end of the comprises five members: T. brucei brucei, T. brucei gambiense, T.
trypanosome and differs in size and position according to the species. brucei rhodesiense, T. evansi and T. equiperdum. The three
It is adjacent to the parabasal body (from which the flagellum arises), subspecies of T. brucei are normally transmitted by tsetse flies (in
and so close to it that it cannot easily be seen separately with the light contrast to T. evansi and T. equiperdum) and are exactly similar in
microscope. The kinetoplast has important functions in reproduction morphology, but only T. brucei gambiense and T. brucei rhodesiense
and metabolism and is probably essential for cyclical transmission by are the cause of human sleeping sickness, the former mainly in West
tsetse flies. (It is sometimes absent in a proportion of. Trypanosomes, and Central Africa and the latter in eastern and southern Africa. T.
especially of some strains of T. evansi, a species which has lost its brucei brucei is not infective to humans.
ability of being cyclically transmitted.) The extent of the undulating
membrane and the absence or presences of the free flagellum are also T. brucei (see Figure 2). T. brucei is polymorphic, with
precious in specific identification of trypanosomes. Other three main forms, all of which have a small kinetoplast and a
morphological characters are the average length and the shape of the conspicuous undulating membrane:
body.

Differential morphology
• Long slender forms (23–30 μm in length) with a free
flagellum, which may be up to one half of the length of the
organism. The posterior end is pointed and the nucleus is
There are distinct differences in appearance, shape and size central. The kinetoplast is placed up to 4 μm in front of the
between the various species of trypanosomes, allowing specific posterior extremity.
identification. It must be remembered, however, that in any biological
• Short stumpy forms (17–22 μm in length) normally without
material there is some variability. In addition, trypanosomes are not
a free flagellum, but in which there may occasionally be
rigid and continuously change their shape slightly; the individual
individuals with a short free flagellum. The kinetoplast is
parasite seen in the stained preparation presents the shape it had now
usually sub terminal. The position of the nucleus varies
of dying. It has also been subjected to the unnatural stresses of drying
greatly and it is in some cases in the posterior part of the
out and being fixed and stained. Many variations in appearance are
cell, sometimes so far posterior that the kinetoplast is
therefore seen. It is thus necessary to observe carefully and
anterior to it (so-called postero-nuclear forms). There is
systematically all the features in a sufficiently large number of
considerable variation in appearance between short stumpy
individual trypanosomes: only after such an examination is it possible
forms, from broad, squat types (which include the postero-
to arrive at a reasonably accurate diagnosis. There will be examples
nuclear forms) to a form similar to T. congolense, although
where trypanosomes are so few, or the staining so inadequate, that
longer. In stained specimens, blue volutin granules are
identification may not be possible or only after a prolonged search. It
often present in the cytoplasm, often arranged in a line
is also essential to examine several individual trypanosomes, because
along the margin of the cell.
even if one specimen is perfect to establish its identity beyond any
doubt, further search may reveal another species and thus a mixed
infection.

4
• Intermediate forms, varying in length between the two
previously mentioned types. A free flagellum, of varying
length, is always present. The nucleus is centrally placed.
The posterior end is somewhat variable in shape, but
usually bluntly pointed. The kinetoplast is close to the
posterior extremity. Volutin granules are occasionally
present but neither as common nor as plentiful as in the
short, stumpy forms.

Trypanosoma brucei blood stream forms Figure 2

The cell structure

The structure of the cell is typical


of eukaryotes, see eukaryotic cell. All major
organelles are seen, including the nucleus,
mitochondria, endoplasmic reticulum, Golgi
apparatus etc. Unusual features include the
single large mitochondria with a condensed
mitochondrial DNA structure, and its
association with the basal body of the
flagellum, unusually the cytoskeleton
organization mechanism of the cell. The cell
surface of the bloodstream form features a
dense coat of variable surface glycoproteins
(VSGs) which is replaced by an equally
dense coat of procyclins when the parasite
differentiates into the procylic in the tsetse
fly midgut.

The genome • 11 pairs of large chromosomes of 1 to 6 megabase pairs.


• 3-5 intermediate chromosomes of 200 to 500 kilobase
The genome of T. brucei is made up of: pairs.

5
• Around 100 mini chromosomes of around 50 to 100 glycoprotein. This cycle is repeated with a pattern of high parasite
kilobase pairs. These may be present in multiple copies per load followed by a period of low parasite load. For this reason,
haploid genome. patients exhibit an irregular pattern of high-grade fevers followed by
a febrile period throughout the course of a systemic infection. For
this reason, vaccine development has been thwarted.
The large chromosomes contain most genes, while the
small chromosomes tend to carry genes involved in antigenic
variation, including the VSG genes. The genome has been sequenced The VSG coat
and is available online.
The surface of the trypanosome is covered by a dense coat
The mitochondrial genome is found condensed into the of ~1x107 molecules of Variable Surface Glycoprotein (VSG). This
kinetoplast, an unusual feature unique to the kinetoplastea class. It coat enables an infecting T. brucei population to persistently evade
and the basal body of the flagellum are strongly associated via a the host's immune system, allowing chronic infection. The two
cytoskeleton structure. properties of the VSG coat that allow immune evasion are:

• Shielding - the dense nature of the VSG coat prevents the


immune system of the mammalian host from accessing the
plasma membrane or any other invariant surface epitopes
The cytoskeleton (such as ion channels, transporters, receptors etc.) of the
parasite. The coat is uniform, made up of millions of copies
The cytoskeleton is predominantly made up of of the same molecule; therefore, the only parts of the
microtubules, forming a sub-pellicular corset. The microtubules lie trypanosome the immune system can 'see' are the N-
parallel to each other along the long axis of the cell, with the number terminal loops of the VSG that make up the coat.
of microtubules at any point roughly proportional to the • Periodic antigenic variation - the VSG coat undergoes
circumference of the cell at that point. As the cell grows (including frequent stochastic genetic modification - 'switching' -
for mitosis) additional microtubules grow between the existing allowing variants expressing a new VSG coat to escape the
tubules, leading to semi conservative inheritance of the cytoskeleton. specific immune response raised against the previous coat.
The microtubules are orientated + at the posterior and - at the
anterior. Microfilament and intermediate filaments also play an Antigenic variation
important role in the cytoskeleton, but these are generally
overlooked.
Sequencing of the T. brucei genome has revealed a huge
VSG gene archive, made up of thousands of different VSG genes. All
Flagellar structure but one of these is 'silent' VSGs, as each trypanosome expresses only
one VSG gene at a time. VSG is highly immunogenic, and an
The trypanosome flagellum has two main structures. It is immune response raised against a specific VSG will rapidly kill
made up of a typical flagellar axoneme, which lies parallel to the trypanosomes expressing this VSG. This can also be observed in
paraflagellar rod, a lattice structure of proteins unique to the vitro by a complement-mediated lysis assay. However, with each cell
kinetoplastida, euglenoids and dinoflagellates. division there is a possibility that one or both of the progeny will
switch expression to a silent VSG from the archive. The frequency of
The microtubules of the flagellar axoneme lie in the normal such a switch has been measured to be approximately 1:100. This
9+2 arrangement, orientated with the + at the anterior end and the - in new VSG will likely not be recognized by the specific immune
the basal body. The cytoskeletal structure extends from the basal responses raised against previously expressed VSGs. It takes several
body to the kinetoplast. The flagellum is bound to the cytoskeleton of days for an immune response against a specific to develop, giving
the main cell body by four specialised microtubules, which run trypanosomes, which have undergone VSG coat switching some time
parallel and in the same direction to the flagellar tubulin. to reproduce (and undergo further VSG coat switching events)
unhindered. Repetition of this process prevents extinction of the
infecting trypanosome population, allowing chronic persistence of
The flagellar function is twofold - locomotion via parasites in the host. The clinical effect of this cycle is successive
oscilations along the attached flagellum and cell body, and 'waves' of parasitaemia (trypanosomes in the blood).
attachment to the fly gut during the procyclic phase.
VSG structure

VSG genes are hugely variable at the sequence level.


Variant Surface Glycoprotein (VSGs) However, for them to fulfill their shielding function, different VSGs
have strongly conserved structural features. VSGs are made up of a
Trypanosomes have a specialized mechanism to overcome highly variable N terminal domain of around 300 to 350 amino acids,
the obstacles of the mammalian immune system. Days after and a more conserved C terminal domain of around 100 amino acids.
infection, host antibodies recognize surface glycoprotein that coat the The C terminal domain forms a structural bundle of 4 alpha helices,
protozoa—the antigenic determinants of the organism—and kill the while the N terminal domain forms a 'halo' around the helices. The
organisms by labeling them destruction. However a few protozoa tertiary structure of this halo is well conserved between different
escape destruction via a programmed system that changes their VSGs (in spite of wide variation in amino acid sequence) allowing
glycoprotein composition and thus enables them to evade immune different VSGs to form the physical barrier required to shield the
recognition. Again, a day later the immune system recognizes this trypanosome's surface. VSG is anchored to the cell membrane via a
change and mounts an immune response against the new glycophosphatidylinositol (GPI) anchor - a covalent linkage from the
C terminus, to approximately 4 sugars, to a phosphatidylinositol
6
phospholipid acid, which lies in the cell membrane. VSGs form Each is a polycistronic unit, containing a number of Expressions Site-
homodimers. Associated Genes (ESAGs) all expressed along with the active VSG.
While there are at least 20 known expression sites, only a single one
VSG archive structure is ever active at one time. A number of mechanisms appear to be
involved in this process, but the exact nature of the silencing is still
unclear
The VSG gene archive is the collection of silent VSGs in
the T. brucei genome. Some of these are full-length, intact genes;
others are pseudogenes) typically with omitted sections or premature The VSG can be switched either by changing the active
stop codons. Expression of an antigenically novel VSG can occur by expression (from the active to a previously silent site) or by changing
simply switching to a different full-length VSG gene. However, only the VSG gene in the active site. The genome contains many copies of
5% of the archive is made up of such complete silent VSGs. To possible VSG genes, both on minichromosomes and in repeated
utilize the rest of the silent VSG archive, ‘mosaic’ VSGs can be sections in the interior of the chromosomes. These are generally
formed by replacing part of the expressed VSG with a structurally silent, typically with omitted sections or premature stop codons, but
homologous region from the archive. The combinatorial nature of are important in the evolution of new VSG genes. It is estimated up
mosaic formation in conjunction with the huge silent VSG archive to 10% of the T.brucei genome may be made up of VSG genes or
gives the parasite a theoretically limitless VSG library, and is the pseudogenes. Any of these genes can be moved into the active site by
major barrier to vaccine development. recombination for expression. Again, the exact mechanisms that
control this are still only partially known.
VSG expression
The Trypanosome cell cycle (procyclic form)
One major focus in trypanosome research is how the
majority of VSG genes are kept silent, and how these genes are The mitotic division of T.brucei is unusual in terms of the
switched. The expressed VSG is always located in an Expression Site cytoskeletal process. The basal body, unlike a centrosome of most
- found at the telomeres of the large and intermediate chromosomes. eukaryotic cells, plays an important role in the organization of the
spindle.

Stages of mitosis:

1. The basal body replicates, both remaining associated with the kinetoplast.
2. The kinetoplast undergoes replication, and the daughter kinetoplasts are separated by the basal bodies.
3. The second flagellum grows while the nucleus undergoes replication.
4. The mitochondrion divides, and cytokinesis progresses from the anterior to posterior end.
5. The division resolves. The daughter cells may stay connected for a significant length of time after cytokinesis is complete

7
The process of endocytosis in trypanosomes lysosomal thiol protease of T. congolense was purified in 1990.
Antibodies to the enzyme have proved to be valuable markers of the
Trypanosomes, like all organisms, need nutrients to live lysosome. An important discovery made during the year was that this
and grow. The parasites obtain these nutrients from their environment enzyme is better recognized by antibodies in the sera of
by taking up molecules and particles in a process known as trpyanotolerant N'Dama cattle recovering naturally from infections
endocytosis. Past studies using the electron microscope have shown than in Zebu cattle that required treatment to recover from infection.
that trypanosomes draw in molecules by way of a structure named the Furthermore, the enzyme is detectable in the sera of cattle early in an
flagellar pocket. Molecules binding to the surface of the infection and so is a possible cause of the immune and physiological
trypanosome, including antibodies, have been found also to be dysfunctions that characterize trypanosomiasis in animals. The
engulfed in this way. Therefore, in addition to changing its surface degree of protection afforded livestock by immunization with the
coat of proteins to avoid antibody binding, the parasite seems able to purified molecule, and the possibility that it is involved in the
take up antibodies, and possibly in this way to neutralize their effect. pathogenesis of the disease, are subjects of current studies.
It was demonstrated during the year that clearance of antibodies from
the surface of the trypanosome in this way renders parasites in vitro Locomotion
resistant to the effects of “complement”, the collective name for a
group of proteins in mammalian blood so named because they Trypanosomes move actively and progress by movement of
complement and amplify the action of antibodies to cause the lysis the undulating membrane and the free flagellum (when present),
(rupture) of parasites. which acts as a kind of propeller, thus drawing themselves through
the blood plasma or tissue fluid. (The free flagellum, when present,
One approach to learning more about the physical arises from the anterior [front] end of the parasite.)
organization of the endocytic pathway is to determine the functional
activity of defined fractions and subcomponents of trypanosomes. In Reproduction
1990, in collaboration with scientists from the European Molecular
Biology Laboratory (Heidelberg), a free-flow electrophoresis
technique was used to separate microsomal trypanosome fractions. This is by a process of division to produce two daughter
The fractions obtained were characterized to some extent based on cells. However, as stated above, it has been shown that exchange and
demonstrations of enzymatic activity. recombination of genetic material may take place in the tsetse fly
between two trypanosomes, but it is unknown how frequently this
occurs.
Progress was also made during the year in purifying and
characterizing a receptor molecule of T. b. brucei that recognizes and
binds to transferring circulating in the blood of the mammalian host. The division into two daughter cells (binary fission)
The receptor appears to be much smaller than its mammalian follows the sequence of events illustrated in Figure 3. The kinetoplast
equivalent. The possibility that trypanosomes have, and make use of, divides first. A second parabasal body develops, from which a second
receptors to mammalian growth factors will be examined in the flagellum develops. The nucleus divides next, followed by the rest of
coming year. the trypanosome body duplicating all the structures present in the
cytoplasm. The body then divides into two daughter cells, beginning
at the anterior end. The process is rapid, and may result in a vast
Parasite enzymes that break down proteins, called population in the host within a short period.
proteases, appear to be involved in the endocytic process. A

8
Division of a trypanosome Fig. 3

Habitat differentiate into several forms, culminating the metacyclic form,


which is able to infect mammalian hosts. When the infected fly next
feeds, these metacyclic trypanosomes are injected into the skin along
• Blood with tsetse saliva. In the animal, the parasites differentiate into a
• Lymph channel throughout the body bloodstream form specially adapted to live in mammalian blood. The
• CSF bloodstream parasites multiply by binary fission and enter the
animal's lymphatic and blood circulation. As flies feed on animals
• Connective Tissue infected with the parasite, they take up blood containing
• Intracellular spaces trypanosomes, which then completes the life cycle.
• Brain
Insects are usually involved in the natural transmission of
Life Cycle the African pathogenic trypanosomes with which we are concerned in
this field guide. When this is the case, the life cycle has two phases,
one in the insect vector and one in the mammalian host. Transmission
The life cycle of the single-celled trypanosome parasite is
by insects may be cyclical by tsetse flies, Glossina species, or
complex. In both the tsetse fly vector and the mammalian host, mechanical by other biting flies (but apart from transmitting
trypanosomes undergo a series of transformations into different
trypanosomes cyclically, tsetse flies can also act as mechanical
forms. The tsetse fly ingests trypanosomes when it feeds on an vectors).
animal infected by the parasite. In the fly, the trypanosomes

9
The tsetse fly is large, brown and stealthy. While taking trypanosomes (metatrypanosomes) are produced. Different
blood from a mammalian host, an infected tsetse fly (genus Glossina) trypanosome species develop in different regions of the digestive
injects metacyclic trypomastigotes into skin tissue. The parasites tract of the fly, and the metatrypanosomes occur either in the biting
enter the lymphatic system and pass into the bloodstream mouthparts or in the salivary glands. The period from ingesting
infected blood to the appearance of these infective forms varies from
one to three weeks; once infective metatrypanosomes are present, the
1. Inside the host, they transform into bloodstream fly remains infective for the remainder of its life. During the act of
trypomastigotes feeding, the fly penetrates the skin with its proboscis. The rupture of
2. are carried to other sites throughout the body, reach other small blood vessels forms a pool of blood in the tissues and the fly
blood fluids (e.g., lymph, spinal fluid), and continue the injects saliva to prevent coagulation. Infection of the host takes place
replication by binary fission at this stage, with infective metacyclic trypanosomes in the saliva.
3. The entire life cycle of African Trypanosomes is
represented by extracellular stages. A tsetse fly becomes Although no classical sexual processes in the life cycle of
infected with bloodstream trypomastigotes when taking a trypanosomes have been described, it has been shown that exchange
blood meal on an infected mammalian host of genomic material (DNA) between trypanosomes sometimes occurs
4. In the fly's midgut, the parasites transform into procyclic in the tsetse fly, although it is not clear how significant this is.
trypomastigotes,
5. multiply by binary fission, Life cycle in the mammalian host. The infective metatrypanosomes
6. leave the midgut, and undergo development and multiplication at the site of infection where
7. transform into epimastigotes a swelling or chancre may be detected in the skin, and finally the
mature blood trypanosomes (or trypomastigotes) are released via
8. The epimastigotes reach the fly's salivary glands and
lymph vessels and lymph nodes into the blood circulation.
continue multiplication by binary fission.

Reproduction in the mammalian host occurs through a


MODE OF TRANSMISSION process of binary division.

Cyclical transmission Trypanosomes feed by absorbing nutrients, through their


outer membrane, from the body fluids of the host. The proteins,
carbohydrates and fats are digested by enzyme systems within their
When a tsetse fly hatches from its pupal case, it is free from
protoplasm. Oxygen dissolved in the tissue fluids or blood plasma of
trypanosomes. Until its first bloodmeal, it is called a teneral fly. It
their host is absorbed in a similar manner, to generate the energy
acquires a trypanosomal infection when feeding on a parasitaemic (=
necessary for the vital processes.
having parasites in the circulating blood) mammalian host. The
trypanosomes undergo a cycle of development and multiplication in
the digestive tract of the fly until the infective metacyclic
10
Waste products are disposed of by a reverse process, By iatrogenic means. This can occur when using the same needle or
through the outer membrane, into the body fluids of the host. They surgical instrument on more than one animal, at sufficiently short
include carbon dioxide formed during respiration, as well as more intervals that the blood on the needle or instrument does not dry. It is
complex metabolic products. not an uncommon occurrence when animals are vaccinated or treated
by injection, or when blood is collected from several animals in a
Life cycle in the tsetse fly. Blood stream forms (trypomastigotes) row, without changing or disinfecting needles or pins. It may also
ingested by the fly undergo considerable changes, in morphology as occur when several animals are subjected at short intervals to a
well as in their metabolism. They change into long slender forms surgical intervention (dehorning, castration, etc.) without properly
called epimastigotes, which multiply and finally give rise to the disinfecting the instruments.
infective meta-trypanosomes.
Transmission by other means
Mechanical transmission
• It is well known that carnivores may be infected with T.
By biting insects. The process is purely mechanical. A biting insect evansi and T. brucei by ingesting meat or organs from
passes the blood forms from an infected animal to another in the infected animals, as long as these are still sufficiently fresh
course of interrupted feeding. The time between the two feeds is to contain live trypanosomes. Infection occurs probably
crucial for effective transmission because the trypanosomes die when through the mucosa of the mouth (in which moreover bone
the blood dries. The importance of this mode of transmission is splinters make wounds through which the parasites
variable from place to place, depending on the numbers of hosts and penetrate even more easily).
biting insects present, and on the species of trypanosome. Large • All trypanosome species are occasionally transmitted
biting insects such as tabanids carry more blood and are more likely congenitally, or vertical transmission from the mother to
to act as mechanical vectors than for example mosquitoes. (Tsetse the offspring, through the placenta either while the fetus is
flies themselves can of course also act as mechanical vectors.) This still in the uterus, or when bleeding occurs during birth.
mode of transmission has proved to be sufficiently effective to Congenital transmission of T. vivax, for example, has been
maintain Trypanosoma vivax and Trypanosoma evansi in South and observed in Latin America as well as in Africa, but its real
Central America, and the latter species in North Africa and Asia as importance is not well known.
well. No tsetse flies occur outside tropical Africa, apart from small
tsetse pockets in the southwest of the Arabian peninsula.

High Risk Group

• Hunters
• Rural villagers living in areas with a lack of infrastructure

 Like running water, electricity

• Workers in game reserves or park


• Tourist who spend an extended time in rural areas or game reserves

11
Disease Course reason for the oscillating levels of parasite load is linked to the ability
of the organisms to change their variable surface glycoproteins
Infection with African trypanosomes can result in disease (VSGs) and evade the host’s immune system. Lymphadenopathy, the
manifestations ranging from asymptomatic or mild to a severe swelling of lymph nodes, especially in the posterior cervical nodes
fulminating disease. T. rhodesiense is more likely to cause a rapidly (on the back of the neck) is characteristic sign of African sleeping
progressing and fulminating disease than T. gambiense. T. gambiense sickness and is termed Winterbottom’s sign.
tends to cause either a slow progressing, which may be self-limiting,
or the central nervous system (CNS). Invasion of the Central Nervous System: Invasion of the central
nervous system (CNS) occurs within several weeks in the Rhodesian
The infection is initiated when metacyclic trypomastigotes species and months to even years in the Gambian species of
are introduced from the saliva of the tsetse into the bite wound. trypanosomiasis. Symptoms include headache, stiff neck, sleep
Generally, there is an asymptomatic incubation period of 1-2 weeks disturbance, and depression, followed by progressive mental
in which the trypomastigotes are replicating within the tissue near the deterioration, focal seizures, tremors, and palsies. This progresses to
site of the bite. Occasionally, a local inflammatory nodule known as a coma and the ultimate death of the patient often secondary to
‘trypanosomal chancre’ is observed during this period. Chancres are pneumonia or sepsis. Without treatment, African trypanosomiasis is
usually tender and painful and ulceration may occur. a universally fatal illness.

The trypomastigotes will invade the capillaries, enter the PATHOGENESIS


circulatory system during this incubation period, and continue to
replicate within the blood of the human host. The establishment of
this acute blood stage infection is characterized by irregular episodes During the bite of an infected tsetse fly, the protozoan
of fever and headache. In the case of T. gambiense the number of enters the bite wound in the fly saliva. The organism multiplies at the
parasites in the blood tends to be very low and often the infected skin site and within a few weeks enters the lymphatics and blood
person exhibits no symptoms, whereas most persons infected with T. circulation. The patient responds with fever and production of IgM
rhodesiense will exhibit much higher parasitemias and a more antibody against the protozoa, and symptoms improve. Within about
pronounced fever sometimes associated with rigor. a week and at roughly weekly intervals thereafter, however, there are
recurrent increases in the number of parasites in the blood. Each of
Disease progression is often characterized by invasion of these bursts of increased parasitemia, meaning parasite in the
the lymphatics in T. gambiense infections. Symptoms during the circulating blood, coincides with the appearance of a new
lymphatic stage include enlarged lymph nodes (particularly post- glycoprotein on the surface of the trypanosomes. More than a
cervical group), weight loss, weakness, rash, itching, and edema as thousand genes, each coding for a different surface glycoprotein, are
well as the continued intermittent febrile attacks. Higher parasitemias present in the protozoan chromosome. Only one of these genes is
are often associated with the symptomatic periods. The infection can activated at a time, and the patient’s immune system must respond to
spontaneously resolve during either the blood stage or the lymphatic each gene product with production of a new antibody. The recurrent
stage. There is usually little evidence of lymphatic involvement in T. cycles of parasitemia and antibody production continue until the
rhodesiense infections. In general, the symptoms during the earlier patient is treated or dies.
stages of the infection tend to be non-specific (fever, malaise,
headache, weakness) and may infect multiple organs. In Trypanosoma brucei rhodesiense infections, the disease
A hallmark feature of African trypanosomiasis is the tends to progress rapidly, with the heart and brain invaded within 6
invasion of the CNS and nervous system impairment. Trypanosomes weeks of infection. Irritability, personality changes and mental
crossing the blood-brain barrier result in a generalized dullness results from involvement, but the patient usually dies from
meningocephalitis characterized by progressively worsening heart failure within 6 months. With Trypanosoma brucei gambiense,
symptoms. Indications of nervous impairment include apathy, progression of infection is much slower, and years may pass before
fatigue, confusion, somnolence, and motor changes (such as tics, death occurs, often from secondary infection. Much of the damage to
slurred speech, and in coordination). The changes in sleep patterns the host is due to immune complexes formed when antibody reacts
are often characterized by extreme fatigue during the day and with complement and high levels of protozoan antigen.
extreme agitation at night. Generally it is 6-12 months (or even years)
after the infection before the neurological symptoms start to become
apparent in the case the CNS stage of the disease will almost always PATHOLOGY
progress to include convulsions or coma by death in both T.
gambiense and T. rhodesiense infections. T. brucei effectively evade the human immune system by a
fascinating genetically programmed system of antigenic variation.
Clinical Presentation When even a single parasite crosses the epithelial layer via the bite of
an infected tsetse fly, local pain and inflammation result as the innate
immune system responds to the invader. Nevertheless, when innate,
Incubation Period: The first clinical manifestation of African
non-specific mechanisms of destruction are not sufficient to clear the
trypanosomiasis occurs a few days after infection as a chancre at the
pathogen (as is often the case in humans), the parasite grows and
site of tsetse fly inoculation. This is due to the localized proliferation
multiples in the bloodstream, while a primary adaptive immune
of the pathogens within the subcutaneous tissue. Incubation period
response is built. Indeed, the adaptive response is evident by the
for T.b. rhodesiense may be two to three weeks, while the incubation
appearance of redness and swelling at the site of infection 1-2 weeks
period for the Gambian species may last several weeks to months.
after the tsetse fly bite. VSGs on the surface of the parasite are
recognized as foreign antigens by specific BCRs and TCRs.
Dissemination: With the conclusion of the incubation period, the Generally, a highly specific B-cell mediated antibody response is
organisms have already disseminated into the bloodstream, leading to generated against to certain VSG epitopes. The adaptive immune
the emergence of a characteristic intermittent fever pattern that response eventually kills all of the clones of the original parasite via
correlates directly with high versus low levels of parasitemia. The
12
antibody-mediated adaptive immune response. However, some United States
parasites spontaneously change the VSG coating by altering which
VSG gen is expression. This process is known as gene conversion. In All cases of African trypanosomiasis are imported from
fact, 806 different genes encode for VSGs. Each VSG gene encodes Africa by travelers to endemic areas. Infections among travelers are
for structurally similar, but extremely unique VSGs. Because rare, with less than 1 case per year reported among US travelers.
different VSGs are distinct enough that they are each seen as Most of these infections are caused by T brucei rhodesiense and are
representative of ‘new’ pathogens by the immune system, a separate, acquired in East African game parks.
primary adaptive immune response must be generated to clear the
parasites with new VSGs. The time required to mount a sufficient
primary adaptive immune response allows the parasite to divide, International
multiply, and occasionally change its VSG surface again. Thus,
although theoretically all T. brucei can eventually be cleared by the African trypanosomiasis is confined to tropical Africa
human immune system, spontaneous changes in which VSG is between latitudes 15°N and 20°S, or from north of South Africa to
expressed allow the parasite to stay one step ahead of the adoptive south of Algeria, Libya, and Egypt.
immune system. Indeed, patients with African trypanosomiasis suffer
from chronic cycles of infection. The disease progresses as these The prevalence of African trypanosomiasis varies by
cycles lead to chronic inflammation, fever, and the build-up of country and region. In 2005, major outbreaks were observed in
immune complexes. Furthermore, the severe and often irreversible Angola, the Democratic Republic of Congo, and Sudan.
neurological damage occurs as parasites reaches deeper and into less
protected areas of the body, such as brain. Fatigue is probably a result
of the body constantly funneling of resources to fight a never-ending In Central African Republic, Chad, Congo, Côte d'Ivoire,
infection. Guinea, Malawi, Uganda, and United Republic of Tanzania, sleeping
sickness remains an important public health problem.

PATHOPHYSIOLOGY Fewer than 50 new cases per year are reported in countries
such as Burkina Faso, Cameroon, Equatorial Guinea, Gabon, Kenya,
Humans are infected with T brucei following a fly bite, Mozambique, Nigeria, Rwanda, Zambia, and Zimbabwe.
which occasionally causes a skin chancre at the site. These injected
trypomastigotes further mature and divide in the blood and lymphatic T. brucei transmission seems to have stopped and no new
system, causing malaise, intermittent fever, rash, and wasting. cases of African trypanosomiasis have been reported for several
Eventually, the parasitic invasion reaches the central nervous system decades in countries such as Benin, Botswana, Burundi, Ethiopia,
(CNS), causing behavioral and neurologic changes such as Gambia, Ghana, Guinea Bissau, Liberia, Mali, Namibia, Niger,
encephalitis and coma. Death may occur. Senegal, Sierra Leone, Swaziland, and Togo.

The parasites escape the initial host defense mechanisms by Sleeping sickness threatens millions of people in 36
extensive antigenic variation of parasite surface glycoproteins known countries of sub-Saharan Africa. The current situation is difficult to
as major variant surface glycoprotein (VSG). This evasion of the assess in numerous endemic countries because of a lack of
humoral immune responses contributes to parasite virulence. During surveillance and diagnostic expertise.
the parasitemia, most pathologic changes occur in the hematologic,
lymphatic, cardiac, and central nervous systems. This may be the
result of immune-mediated reactions against antigens on red blood In 1986, a panel of experts convened by the World Health
cells, cardiac tissue, and brain tissue, resulting in hemolysis, anemia, Organization (WHO) estimated that 70 million people lived in areas
pancarditis, and meningoencephalitis. where transmission of African trypanosomiasis is possible. In 1998,
almost 40,000 cases of the disease were reported, but this number did
not reflect the true situation given the remoteness of affected regions
A hypersensitivity reaction causes skin problems, including and the focal nature of the disease. Between 300,000 and 500,000,
persistent urticaria, pruritus, and facial edema. Increased lymphocyte more cases were estimated as remaining undiagnosed and therefore
levels in the spleen and lymph nodes infested with the parasite leads untreated.
to fibrosis but rarely hepatosplenomegaly. Monocytes, macrophages,
and plasma cells infiltrate blood vessels, causing endarteritis and
increased vascular permeability. During recent epidemic periods, the prevalence of sleeping
sickness has reached 50% in several villages in the Democratic
Republic of Congo, Angola, and Southern Sudan. Sleeping sickness
The gastrointestinal system is also affected. Kupffer cell was considered the first or second greatest cause of mortality in those
hyperplasia occurs in the liver, along with portal infiltration and fatty communities, even ahead of HIV infection and AIDS. By 2005,
degeneration. Hepatomegaly is rare. More commonly in East African surveillance had been reinforced and the number of new cases
trypanosomiasis, a pancarditis affecting all heart tissue layers reported throughout the continent had substantially reduced; between
develops secondary to extensive cellular infiltration and fibrosis. 1998 and 2004, the figures for both forms of African trypanosomiasis
Arrhythmia or cardiac failure can cause death prior to the together fell from 37,991 to 17,616.
development of CNS manifestations. CNS problems include
perivascular infiltration into the interstitium in the brain and spinal
cord, leading to meningoencephalitis with edema, bleeding, and The estimated number of cases is currently between 50,000
granulomatous lesions. and 70,000. The current epidemic, which began in 1970, is thought to
have been facilitated by factors such as the halting of screening
programs, population migration, civil war, economic decline, and
FREQUENCY reduced health care financing.

13
Mortality/Morbidity Cardiovascular alterations are less prominent, especially in
the Gambian form. Irregular febrile episodes are accompanied by
headaches, malaise, exhaustion, anorexia, extreme thirst, muscle and
• The symptoms of East African trypanosomiasis develop
joint pains, pruritus, anaemia, rash and often-deep hyperesthesia (the
more quickly (starting 1 month after bite) than the
sign of the key of Kerandel). The lymph nodes are generally rubbery
symptoms of West African trypanosomiasis, which can
and mobile, painful at the beginning. Palpation of the subclavicular
begin months to a year after the first bite.
region (Winterbottom sign) is an important part of the diagnostic
• Both types of African trypanosomiasis cause the same procedure in the Gambian form. Any adenopathy accompanied by
generalized symptoms, including intermittent fevers, rash, fever should evoke the diagnosis of sleeping sickness in patients from
and lymphadenopathy. Notably, individuals with the East endemic areas. Later, pruritus generalizes. Edema of the face and
African form are more likely to experience cardiac extremities appears early.
complications and develop CNS disease more quickly,
within weeks to a month. The CNS manifestations of
behavioral changes, daytime somnolence, nighttime Few minor neurological and endocrine disorders may
insomnia, stupor, and coma result in death if untreated. reveal the precocity of central nervous system (CNS) involvement,
long before any detectable changes occur in the cerebrospinal fluid
• In West African trypanosomiasis, the asymptomatic phase (CSF). Daytime somnolence or nighttime insomnia may already be
may precede onset of fevers, rash, and cervical reported and electroencephalographic (EEG) tracings may reveal
lymphadenopathy. If unrecognized, the symptoms then abnormalities. Psychiatric signs, with the alternation of irritability,
progress to weight loss, asthenia, pruritus, and CNS disease changes in personality or mood affecting the daily and professional
with a more insidious onset. Meningismus is rare. Death at life of the patients, constitute often the first manifestation of the
this point is usually due to aspiration or seizures caused by disease. A permanent feeling of coldness marks the endocrine
CNS damage. syndrome, lack of appetite or in contrast hyperphagia, polydipsia and
impotence, amenorrhea or infertility, indicative of vegetative and
Race sexual disturbances.

African trypanosomiasis has no racial predilection. Stage 2, The Meningoencephalitic Stage

Sex The meningoencephalitic stage appears slowly and


insidiously over a period of months or years depending on the
African trypanosomiasis has no sexual predilection. trypanosome. However, the clinical signs remain reversible for a long
time with treatment attesting to the predominance of potentially
reversible inflammatory lesions over irreversible demyelinating
Age lesions. The general signs of the haemolymphatic stage do not
completely disappear: spikes of fever (but sometimes hypothermia),
Exposure can occur at any time. Congenital African adenopathies and splenomegaly, cardiovascular manifestations
trypanosomiasis occurs in children, causing psychomotor retardation endocrine disturbances and typical pruritus.
and seizure disorders.
The development of the neurological symptoms is
CLINICAL SYMPTOMATOLOGY OF HUMAN AFRICAN progressive. As neurological signs occur already in stage I, biological
criteria are the only means to confirm CNS invasion. The threshold
TRYPANOSOMIASIS criteria, which are commonly used, are based on CSF examination:
more than 5 cells/µL and/or the presence of trypanosomes.
The symptoms of two diseases are more pronounced in
Caucasian than in the local African population. A wide variety of symptoms is encountered. The main
symptoms from which sleeping sickness was named are daytime
After a painful tsetse bite, the chancre represents the initial somnolence and nocturnal insomnia, the patients being "sleepy by
lesion at the bite site, characterized by local erythema, edema, heat, day and restless by night". The sleep-wake cycle disturbances are
tenderness and a lack of any suppuration. Trypanosomes are present accompanied with either or many of the following symptoms:
in the inflammatory tissues. The chancre disappears within 2 or 3 headaches, sensory disturbances with diffuse superficial or deep
weeks. The disease evolves in two distinct successive phases sensations (muscle and bone hyperesthesia, either spontaneous or
determining its two pathological stages. Within a few days after the provoked; hyperpathia), presence of primitive reflexes (palm-mental
tsetse bite, the patient enters the haemolymphatic stage I of the reflex, sucking reflex), exaggerated deep tendon reflexes, psychiatric
illness. disorders (confusion, mood swings, agitation, aggressive behaviour,
euphoria, absent gaze, mutism, indifference), and tremor (fine and
diffuse without any myoclonic jerk at rest or during movement).
Stages 1, The Haemolymphatic Stage Pyramidal alterations revealed by a Babinski sign can also be
observed along with alterations in muscle tone, numbness or sensory
Clinical signs appear very early. Intermittent fever develops deficit.
because of the successive waves of invasion of the blood by the
trypanosomes. Adenopathies, splenomegaly, or even hepatological An abnormal number of monocytic cells is observed in the
signs mark the invasion of the reticulo-endothelial system. Skin CSF. The early neurological symptoms correlate with the widespread
eruptions or trypanides are commonly observed. Severe pruritus with meningeal inflammation, which occurs in both forms of HAT. The
scratching skin lesions becomes unsupportable for the patient. selective CNS locations explain in part the principal clinical
neurological signs. Sleep-wake disturbances may result from invasion

14
of the median eminence by a parasite, which also accounts for neuro- with West African trypanosomiasis. The classic
endocrine dysfunctions with the involvement of the suprachiasmatic Winterbottom sign is clearly visible (ie, enlarged,
nuclei. Disorders of the sleep-wake cycle are accompanied by a state nontender, mobile posterior cervical lymph
of apathy in the patient, the loss of muscle tone especially in the neck node).
muscles and a drooping of the eyelids. Extrapyramidal symptoms o Fevers, tachycardia, irregular rash, edema, and
signal the involvement of the striatum. Deep sensory disturbances weight loss
with hyperpathia may result from the involvement of the thalamus o Organomegaly, particularly splenomegaly (T
and the early invasion of posterior spinal roots. brucei gambiense African trypanosomiasis)
• Stage 2 (late, or CNS, stage)
Apart from the disruptions of the circadian rhythm of the o CNS symptoms: The CNS symptoms of West
sleep-wake cycle, other biological rhythms are disturbed, such as African trypanosomiasis have a slower onset of,
body temperature, cortisol and prolactin or growth hormone ie, months to a year. Symptoms include
secretion. The invasion of the subthalamic and hypophyseal regions irritability, tremors, increased muscle rigidity and
account for the persistence of ad-endocrine disturbances such as tonicity, occasional ataxia, and hemiparesis, but
impotence, amenorrhea or infertility and the development of rarely overt meningeal signs. East African
disturbed sensations of hunger and thirst, with often hyperphagia and trypanosomiasis usually has a faster onset, ie,
polydipsia in contrast to the poor general state of malnutrition of the weeks to a month, and does not exhibit a clear
patients. At the terminal phase of the disease, CNS demyelination and distinction between the two stages.
atrophy are accompanied with disturbances in consciousness and the o Kerandel sign, including delayed pain on
development of dementia with incoherence, incontinence and compression of patient's soft tissue
epileptic fits. The patient dies in a state of cachexia and physiological o Behavioral changes consistent with mania or
misery. psychosis, speech disorders, and seizures
o Stupor and coma (giving rise to the name
History sleeping sickness)
o Psychosis
• Stage 1 (early, or hemolymphatic, stage) o Sensory disorders, tremor, and ataxia
o Painless skin chancre that appears about 5-15
days after the bite, resolving spontaneously after Disease Management
several weeks (seen less commonly in T brucei
gambiense infection) Disease management is performed in three steps:
o Intermittent fever (refractory to antimalarials),
• Screening for potential infection. This involves the use of
general malaise, myalgia, arthralgias, and
serological test and/or checking for clinical signs –
headache, usually 3 weeks after bite
generally swollen cervical glands.
o Generalized or regional lymphadenopathy
(Posterior cervical lymphadenopathy • Diagnosing shows whether the parasite is present.
[Winterbottom sign] is characteristic of T brucei • Staging to determine the state of progression of the disease
gambiense African trypanosomiasis [sleeping entails examination of cerebro-spinal fluid obtained by
sickness].) lumbar puncture and is used to determine the course of
o Facial edema (minority of patients) treatment.
o Transient urticarial, erythematous, or macular Diagnosis must be made as early as possible and before the
rashes 6-8 weeks after onset neurological stage in order to avoid complicated, difficult and risky
o Trypanids (ill-defined, centrally pale, evanescent, treatment procedure.
annular or blotchy edematous erythematous
macules on trunk)
• Stage 2 (late, or CNS, stage) DIAGNOSIS
o Persistent headaches (refractory to analgesics)
o Daytime somnolence followed by nighttime
insomnia Laboratory Studies
o Behavioral changes, mood swings, and, in some
patients, depression • General
o Loss of appetite, wasting syndrome, and weight o In African trypanosomiasis (sleeping sickness),
loss the most significant laboratory abnormalities
o Seizures in children (rarely in adults) include anemia, hypergammaglobulinemia, low
complement levels, elevated erythrocyte
Physical sedimentation rate (ESR), thrombocytopenia, and
hypoalbuminemia, but not eosinophilia or
abnormal liver function.
• Stage 1 (early, or hemolymphatic, stage) o In West African trypanosomiasis, the total
o Indurated chancre at bite site immunoglobulin M (IgM) level is notably higher
o Skin lesions (trypanids) in light-skinned patients in blood and CSF (along with high CSF protein).
o Lymphadenopathy: Axillary and inguinal o A definitive diagnosis of infection requires actual
lymphadenopathy are more common in patients detection of trypanosomes in blood, lymph nodes,
with East African trypanosomiasis. Cervical CSF, skin chancre aspirates, or bone marrow.
lymphadenopathy is more common in patients However, symptomatic improvement after
15
empiric treatment is the usual confirmatory test in • General: Field serology-based diagnosis of African
areas where diagnostic studies are not readily trypanosomiasis has been slow to progress over the past
available. decades. Although many research tools are available for
• Lymph node aspiration at a high dry magnification (X400) diagnosis, few are used clinically in endemic areas.
is commonly used as a rapid test for trypanosomes. It • Serologic antibody detection
requires immediate search for parasites because they are o The standard serologic assay to diagnose West
mobile for only 15-20 minutes. This test has more utility in African trypanosomiasis is the card agglutination
T brucei gambiense trypanosomiasis. test for trypanosomiasis (CATT).
• Blood smear o The CATT can be conducted in the field without
o A wet smear of unstained blood or Giemsa- electricity, and results are available in only 10
stained thick smear (more sensitive) is used to minutes. It is highly sensitive (96%) but less
evaluate for mobile trypanosomes, again for 15- specific because of cross-reactivity with animal
20 minutes. Wright and Leishman stains are trypanosomes.
inadequate. This technique is most sensitive in o Commercial antibody tests for Eastern African
early stages of disease, when the number of trypanosomiasis are not available.
circulating parasites is highest (≥5000/mL), • Antigen detection tests based on enzyme-linked
particularly in T brucei rhodesiense immunosorbent assay (ELISA) technology have been
trypanosomiasis. developed. They have shown inconsistent results and are
o Better assays are now available, including the not yet commercially available.
hematocrit centrifugation technique for buffy
• Culture of CSF, blood, bone marrow aspirate, or tissue
coat examination and the miniature anion-
specimens can be performed in liquid media.
exchange centrifugation technique (mAECT),
which filters out the red cells but not the • Other tests developed but not frequently used clinically
trypanosomes. This test can be used to detect include antibody detection in the CSF and intrathecal space
parasitemia levels as low as 5 parasites/mL; the (low sensitivity), polymerase chain reaction (PCR), and
test can be repeated on subsequent days to serum proteomic tests.
increase the yield when results are negative. • Research tools such as isoenzyme analysis and restriction
• Chancre aspirate can be used as a wet preparation, fragment length polymorphism (RFLP) are used for
especially in East African trypanosomiasis, but a blood definitive subspecies identification.
smear is more sensitive.
• Bone marrow aspiration results may be positive in some Procedures
patients.
• CSF assay • Lumbar puncture: CSF fluid is used to detect trypanosomes
o Lumbar puncture should be performed whenever and to measure WBC counts, protein, and IgM in patients
trypanosomiasis is suspected. CSF examination with parasitemia or positive serologies or symptoms.
helps to diagnose and stage the disease. However, Importantly, CNS disease can manifest early in East
a negative result does not necessarily rule out the African trypanosomiasis.
diagnosis.
o The double centrifugation technique is the most Differential Diagnoses
sensitive method to detect the trypanosomes.
o Other CSF findings include elevated WBC count,
elevated IgM levels, elevated total protein levels, Other Problems to Be Considered
and raised intracranial pressure. An uncommon
characteristic finding is Mott cells, which are • Stage 1 (early) African trypanosomiasis (sleeping sickness)
thought to be large eosinophilic plasma cells Symptoms
containing IgM that have failed to secrete their
antibodies.
o Increased intrathecal synthesis of IgM has been Differential diagnoses of recurrent fever include malaria,
HIV infection, borreliosis, and brucellosis, typhoid fever, and other
found to be the most sensitive indicator of CNS
enteric fevers.
involvement in African trypanosomiasis.

Differential diagnoses of lymphadenopathy include


tuberculosis (TB) lymphadenitis, HIV infection, and cancer.

Imaging Studies • Stage 2 (late) African trypanosomiasis symptoms

• CT scanning and MRI of the head: Both head CT scanning Differential diagnoses of mental status changes include TB,
and MRI reveal cerebral edema and white matter meningitis, and HIV-related opportunistic infections, including
enhancement, respectively, in patients with late-stage cryptococcal meningitis.
African trypanosomiasis.
• EEG in neurologic involvement usually shows slow wave
oscillations (delta waves), a nonspecific finding. TREATMENT

Other Tests Medical Care

16
• Prehospital care of African trypanosomiasis (sleeping Medication
sickness) centers on management of the acute symptoms of
fever and malaise while closely monitoring the patient’s The type of drug treatment used depends on the type and
neurologic status. stage of African trypanosomiasis (sleeping sickness).The drugs used
• In the emergency department, if CNS symptoms are severe, in the first stage of the disease are less toxic, easier to administer and
then airway management to prevent aspiration becomes more affective. Treatment success in the second stage defends on a
important, along with an immediate blood smear, CBC drug that can cross the blood –brain barrier to reach the parasite.
count, and lumbar puncture for trypanosome detection. Such drugs are quite toxic and complicated to administer. Four drugs
are registered for the treatment of sleeping sickness and provided free
of charge to endemic countries through a WHO private partnership
with anofi-aventis (pentamidine, melarsoprol, eflornithine) and Bayer
AG (suramin).

Medications Medications
Stage 1 Stage 2
Type of Trypanosomiasis
(Hemolymphatic Stages) (Neurologic [CNS] Stages)

East African trypanosomiasis


Suramin 100-200 mg IV test dose, then Melarsoprol 2-3.6 mg/kg/d IV for 3d;
(caused by T brucei rhodesiense)
1 g IV on days 1,3,7,14,21 after 1 week, 3.6 mg/kg/d for 3days; after
10-21 d, repeat the cycle

West African trypanosomiasis


Pentamidine isethionate 4 mg/kg/d IM Melarsoprol 2-3.6 mg/kg/d IV for 3d;
(caused by T brucei gambiense)
for 10 d or after 1 week, 3.6 mg/kg/d for 3 days;
after 10-21 d, repeat the cycle or
Suramin 100-200 mg IV test dose, then
1 g IV on days 1,3,7,14,21 Eflornithine 400 mg/kg/d IV in 4 divided
doses for 14 days

Suramin (Metaret) during the grand epidemic in West and Central Africa in millions of
people and was the mainstay of therapy until 1969.
Antiparasitic agent used IV in early-stage African
trypanosomiasis and onchocerciasis. Suramin is a polysulfonated Adult
naphthylamine derivative of urea. Suramin is trypanocidal and works
by inhibiting parasitic enzymes and growth factors. Highly bound to 100-200 mg test dose, then 1 g IV on days 1, 3, 7, 14, 21
serum proteins and, thus, crosses the blood-brain barrier poorly.
Serum levels are approximately 100 mcg/mL. Suramin is more
effective and less toxic than pentamidine. Excreted in the urine at a Pediatric
slow rate.
1-2 mg test dose, then 20 mg/kg IV on days 1, 3, 7, 14, 21
Suramin was introduced in 1920 to treat the first stage of
the disease. By 1922, Suramin was generally combined with Melarsoprol (Melarsen Oxide-BAL, Mel B, RP 3854)
Tryparsamide (another pentavalent organo-arsenic drug) in the
treatment of the second stage of the gambiense form. It was used Trivalent arsenical used in the late or CNS stage of African
trypanosomiasis. Trypanocidal, inhibiting parasitic glycolysis. Water
17
insoluble and has a half-life of 35 h. Serum levels range from 2-5 enzyme, thereby interfering with parasite aerobic glycolysis. Because
mcg/mL, but CSF levels are 50-fold lower. The kidneys primarily of poor GI absorption, the drug is administered IV/IM and is strongly
excrete the drug. Clinical improvement is usually observed within 4 d bound to tissues, including spleen, liver, and kidney. Clinical
after starting the drug. Therapy is as high as 90-95% successful in improvement usually noted within 24 h of injection. Reported to have
clearing the parasitemia. However, it can be toxic and even fatal in 4- a >90% cure rate. Pentamidine does not penetrate the blood-brain
6% of cases. Studies have now demonstrated the effectiveness of 10- barrier effectively and, therefore, does not treat CNS infection.
day melarsoprol treatments for late-stage African trypanosomiasis. In
addition, melarsoprol resistance has become a concern in the Congo Pentamidine, a highly effective drug for the first stage of
and Uganda; up to 30% of cases do not respond to the drug. the disease, has been used since 1939. During the fifties, it was
widely used as a prophylactic agent in Western Africa, leading to a
The organo-arsenical melarsoprol (Arsobal) was developed sharp decline in infection rates. At the time, it was thought that
in the 1940s, and is effective for patients with second stage sleeping eradication of the disease was at hand.
sickness. However, 3 - 10% of those injected have reactive
encephalopathy (convulsions, progressive coma, or psychotic Adult
reactions), and 10 - 70% of such cases result in death; it can cause
brain damage in those who survive the encephalopathy. However,
due to its effectiveness, melarsoprol is still used today. Resistance to 4 mg/kg/d IM/IV for 10 d
melarsoprol is increasing, and combination therapy with nifurtimox is
currently under research. Pediatric

Adult Administer as in adults

2-3.6 mg/kg/d IV for 3 d; after 1 wk, 3.6 mg/kg/d IV for 3 d; after 10- During the first half of the 20th century, there were very few
21 d, repeat the cycle; always administer each dose slowly on an efficacious medications. The only one, which was known and used at
empty stomach first, was an arsenical by-product named Atoxyl it had to be
administered over a several week period of time through course of
Pediatric subcutaneous injection. As it was a swift killer of trypanosomes
roaming the blood stream, its use during the first stage prevented the
risk of transmission with new fly bites. However, as this medication
0.36 mg/kg IV initially; increase gradually to a maximum 3.6 mg/kg does not pass through the meningeal barrier it is not efficacious
at intervals of 1-5 d for a total of 9-10 doses; average dosing is 18-25 against trypanosomes which are already present in the cerebro-spinal
mg/kg over 1 month fluid so that the disease can continue to progress in patients who are
experiencing the second stage of this condition.
Eflornithine (Ornidyl)
In 1926, a new arsenical by-product named Tryparsamide
Recommended for treatment of patients with West African became available. It is given intravenously once a week over a few
trypanosomiasis, especially late (or CNS) disease. Selective and weeks. This medication has a great advantage over Atoxyl as it
irreversible inhibitor of ornithine decarboxylase, which is a critical penetrates into the cerebro-spinal fluid. It is therefore a medication
enzyme for DNA and RNA synthesis. Generally tolerated better and for the second stage of the disease. Nevertheless about 20% of all
is less toxic than arsenic drugs. Available via World Health patients are not going to recover and their outcome will be fatal one.
Organization. Initial response time is 1-2 wk, used for patients in
whom melarsoprol fails. When mass therapy campaigns against Tyrpanosomiasis
got started, only these two medications were available. They are
Eflornithine (difluoromethylornithine or DFMO), the most arsenical by-products with deleterious side effects for the optic nerve,
modern treatment, was developed in the 1970s by Albert as a result, most of the time patients are cured but some complain of
Sjoerdsmanot and underwent clinical trials in the 1980s. The drug ocular side effects and may even wind up fully blind. The need of
was approved by the United States Food and Drug Administration in repeated injections is another shortcoming as it is difficult to have
1990, but Aventis, the company responsible for its manufacture, patients stick to the program unless they are inpatient over two or
halted production in 1999. In 2001, however, Aventis, in association three month period. Then incomplete treatment course give rise to
with Médecins Sans Frontières and the World Health Organization, strains which are resistant to these medication. These first generation
signed a long-term agreement to manufacture and donate the drug. arsenical drugs are obsolete now and not used any longer with the
availability of new molecules, which were developed through
Adult research and development.

100 mg/kg IV q6h for 14 d An international research team working in the Democratic
Republic of the Congo, New Sudan and Angola involving Immtech
International and University of North Carolina Hill has completed a
Pediatric (Not established) Phase IIb clinical trial and commenced a Phase III trial in 2005
testing the efficacy of the first oral treatment for Sleeping Sickness,
Pentamidine isethionate (Pentam 300, Pentacarinat, NebuPent) known at this point as “DB289”.

Antiprotozoal agent usually used for early (or stage 1)


African trypanosomiasis as well as Pneumocystis carinii pneumonia
and leishmaniasis. Works by inhibiting dihydrofolate reductase
18
Combination therapy • In late, or stage 2, trypanosomiasis, the CNS manifestations
are ultimately fatal if untreated. The cure rate approaches
Combination therapy may be more effective than 95% with drugs that work inside the CNS (eg,
monotherapy for the treatment of late-stage T brucei gambiense melarsoprol).
trypanosomiasis. An open randomized trial involving 278 patients
compared melarsoprol monotherapy (two different dosing regimens),
nifurtimox monotherapy, and melarsoprol-nifurtimox combination
MEDICAL ECOLOGY OF SLEEPING SICKNESS: THE
therapy. The 48 relapses reported in the study were limited to patients VECTOR
receiving one of the 3-monotherapy regimens. The trial concluded
that a consecutive 10-day low-dose melarsoprol-nifurtimox
Distribution
combination is more effective than the standard melarsoprol regimen.

Tsetses (pronounced se’tse’, tse’tse’), sometimes-spelled


A trial that compared the efficacy of eflornithine
tzetze, are large biting flies inhabiting much of mid-continental
monotherapy to nifurtimox-eflornithine combination therapy in
Africa between the Sahara and Kalahari deserts. They live by feeding
patients with late-stage T brucei gambiense infection found that cure
on the blood of vertebrate animals and are the primary biological
rates were similar. However, adverse effects were more common in
vectors of trypanosomes, which causes human sleeping sickness and
patients who received eflornithine monotherapy (25.5% vs 9.6%).
animal trypanosomiasis, called nagana.
Thus, the nifurtimox-eflornithine combination appears to be a
promising regimen for use in late-stage T brucei gambiense
trypanosomiasis. Tsetse flies are 6-14 mm long, excluding the proboscis. The
broad head bears three ocelli and two large compound eyes, which
are widely spaced in both sexes. The pair of short antennae has three
Anthelmintics
segment and each an arista with its characteristically branched hairs.
Hygro-, thermo-, chemo- and mechanoreceptors are concentrated on
Parasite biochemical pathways are sufficiently different the antennae. The mouthparts consist of three unpaired components,
from the human host to allow selective interference by the piercing labium, the labrum enclosing the food channel, and the
chemotherapeutic agents in relatively small doses. hypopahrynx with the salivary channel. All are sheathed by two
modified maxillarypalps. On the back, the thorax ends in a caudal,
Follow-up triangular scutellum. The eight abdominal segments can distend
greatly after feeding. Genital organs, males possessing a button-like
hypopygium under the dorsal end of the body, which covers the
Further Inpatient Care
clasper, can only distinguish males and females. The white larvae are
elongated, without eyes and legs, at the posterior end possessing a
• If late or CNS disease complications and coma occur, pair of black polypneustic lobes.
intensive care unit (ICU) staff are needed while treatment is
administered (i.e., melarsoprol for East African There are 31 species and subspecies of tsetse flies under the
trypanosomiasis or eflornithine for West African genus Glossina, family Glossinidae, and order Diptera. Tsetse flies
trypanosomiasis). Monitor potential adverse effects from are largely classified into three subgenera based on morphological
such drugs, including hematologic, renal, and hepatic differences in the structure of the genitalia: Morsitans (Glossina),
function. Palpalis (Nemorhina), and Fusca (Austenina) groups. Although the
tsetse flies can be found over some 9 million squared kilometers of
Further Outpatient Care the African continent, presence of glossina populations throughout
the continent are far from continuous. In general, the Sahara and
Somali Deserts limit the populations in the north, extending across
• In both early- and late-stage trypanosomiasis, treatment the entire continent from Senegal in the west to southern Somalia in
usually resolves symptoms and clears the parasitemia on the east. Tsetse populations are denser in West and Central Africa,
repeat blood smears. and are found more sporadically to the East and down to the borders
• Patients who have recovered from late-stage East African of the Kalahari and Namibian Deserts in Southern Africa. Although
trypanosomiasis should undergo lumbar punctures every 3 tsetse fly habitats may vary considerably, climate and altitude -
months for the first year. Patients who have recovered from through their direct effects on vegetation, rainfall, and temperature -
West African trypanosomiasis should undergo lumbar are still the primary determinants for proliferation. Unlike other
punctures every 6 months for 2 years. A relapse is insects, there are no seasonal interruptions in the life cycles of tsetse
suggested if symptoms return, the CSF WBC count is flies. However, both adult longevity and puparial duration are related
above 20 cells/µL, CSF pleocytosis occurs, or if to temperature, and a significant seasonal decline in tsetse
trypanosomes are still present in blood or CSF. A populations is normal, particularly in savannah habitats during the
persistently elevated CSF WBC count can be observed in dry season.
recovering patients, however, so the change (increase or
decrease) in WBC count is more helpful diagnostically. If a
Behavioral Ecology of the Vector: Mating
relapse is noted, consider repeat treatment with melarsoprol
or eflornithine.
The mating behaviors of tsetse flies have received much
attention because of the development of the Sterile Insect Technique
Prognosis
(SIT) for tsetse fly control. The existences of tsetse flies at low
densities in certain areas suggest highly specific mating mechanisms
• In early, or stage 1, trypanosomiasis, most patients involving visual and olfactory responses. Female tsetse flies only
experience full recovery following treatment. need to mate once in their life time, but multiple mating have been
19
known occur occasionally. Cross mating is possible in areas where currently under surveillance, and relatively few cases are accurately
habitats of different species overlap, however male hybrids are diagnosed annually. Although sleeping sickness was largely under
infertile. Mating of female is mostly confines to early life with mean control during the 1960s, recent epidemics have been strongly
duration of mating declining by age. Most female flies are associated with political and civil unrest in West and Central Africa
successfully inseminated even at very low population densities; resulting in mass movement of populations into areas formerly
usually during their first blood meal right after emerge from the pupal uninhabited by humans.
stage.
In 2005, major outbreaks have been observed in Angola,
Behavioral Ecology of Vector: Host Feeding the Democratic Republic of Congo and Sudan. In Central African
Republic, Chad, Congo, Côte d’Ivoire, Guinea, Malawi, Uganda and
Tsetse flies use visual and olfactory characteristics to United Republic of Tanzania sleeping sickness remains an important
recognize potential hosts before initiating host-oriented responses. public health problem. Countries such as Burkina Faso, Cameroon,
There are series of behavioral responses involved in the process of Equatorial Guinea, Gabon, Kenya, Mozambique, Nigeria, Rwanda,
obtaining a blood meal. Host-seeking behaviors are influenced by Zambia and Zimbabwe are reporting fewer than 50 new cases per
endogenous and exogenous factors. Endogenous factors include year. In countries such as Benin, Botswana, Burundi, Ethiopia,
circadian rhythm of activity level of starvation, age, sex, and Gambia, Ghana, Guinea Bissau, Liberia, Mali, Namibia, Niger,
pregnancy status of the fly. Exogenous factors include temperature, Senegal, Sierra Leone Swaziland and Togo transmission seems to
vapor pressure, visual and olfactory stimuli, and mechanical have stopped and no new cases have been reported for several
stimulation. decades. Nonetheless, it is difficult to assess the current situation in a
number of endemic countries because of a lack of surveillance and
diagnostic expertise.

Four Stages of Host-locating Behavior Epidemiology of West African Sleeping Sickness

• Ranging Both protozoa species are morphologically


Flying in search of a host in the absence of an indistinguishable but have drastically different epidemiological
external cue. features
• Activation
Change in behavior caused by perception of West African sleeping sickness is typically a chronic
external stimulus disease, making it a difficult disease to diagnose in the field. Low
levels of trypanosomes in circulating blood make it difficult to detect
• Orientation
the presence of parasites in blood smears, requiring more
Upwind anemotaxis in response to complex
sophisticated means of detecting trypanosomes such as with the use
chemical and visual directing the insect to the
of miniature anion-exchange/centrifugation (mAEC) technique. In
host
comparison to the East African form, T. b. gambiense has a longer
• Landing evolutionary history with humans, having successfully adapted to
Generally, the tsetse fly will detect an odor plume establishing infections in human hosts without manifesting severe
upwind until it visually recognizes the host. After symptoms. Astonishingly, infection rates of T. b. gambiense in wild
landing on the host, heat stimulation cause a glossina populations are as low 0.1%, even in areas with an epidemic
probing and feeding response. of sleeping sickness.

Life Cycle of the Vector Vectors of West African sleeping sickness are species of
the palpalis group, most of which are in close contact with humans.
Unlike most insect species (r-strategists) that produce large Several different reservoirs for T. b.gambiense have been identified,
quantities of eggs, fertilized female tsetse flies (k-strategies) “give strongly suggesting that other animals, such as the African domestic
birth” to one larva. A typical female tsetse fly will produce one full pig, may maintain the persistence of sleeping sickness in human
grown larva approximately every 9-10 days depending on the populations. However, T.b.gambiense has not been observed or
temperature and humidity. A single egg will hatch and develop to a proven experimentally to reach significantly infectious levels of
third-stage larva in the uterus of the female fly, where it is nurtured parasetemia in other reservoir host. Although it is widely accepted
and supplied with nutrients. This reproductive process is known as that the human-to-fly contact is the main route of transmission, some
adenotrophic viviparity. suggest a minor cycle involving an animal reservoir may help explain
the re-emerge and persistence of the disease in West Africa.
MEDICAL ECOLOGY OF SLEEPING SICKNESS: THE The epidemiology of T.b. gambiense sleeping sickness is
HUMAN far from being fully understood. Despite the low levels of parasitemia
in humans, the disease has successfully established endemicity in
Although epidemics as large as the ones in Uganda at the many regions of West Africa. It has also long been observed that the
turn of the century have not been repeated, there is much concern incidence of disease is not related to the density of the glossina
over the re-emergence and increase in the number of sleeping populations, and that epidemics often occur in areas where the
sickness cases being reported every year in Africa. In 1994, there density of the vector is low. In Nigeria, sleeping sickness occurred in
were an estimated 150,000 cases in Congo, with prevalence as high the north where the distribution of G.p. palpalis and G.tachinoides
as 70% in some villages. Despite the WHO projection of 60 million were scare and restricted to vegetation close to watercourses during
people at risk in Africa, only a fraction of the population at risk is the dry season. In Southern Nigeria, the same species of tsetse flies
are found in abundance due to favorable climatic conditions, yet
20
cases of sleeping sickness have never been observed. It is thought effective in preventing other insects from biting and
that the nature of the human-fly contact is of particular importance in causing illness.
the transmission of T. b. gambiense and the distribution of the
disease, and that human-fly contacts can be classified as “personal” Control
or “impersonal” depending on the ecological circumstances of the
interaction.”Personal” contact refer to situations where fly
movements are restricted to areas where exposure to humans are In the absence of vaccine for trypanosomiasis and the
frequent, such as a watering hole or a stream, and single tsetse fly can looming threat of further trypanocidal drug resistance, the most
have multiple opportunities to feed on humans. “Impersonal” contact theoretically desirable means of controlling the disease is through the
occurs when fly movements are less restricted, and where repeated vector population. Although complete eradication of the vector is
contacts are not likely. In general, ecological isolation of tsetse flies impossible, the most successful attempts at controlling tsetse flies are
in the vicinity human populations leads to increased “personal” likely to be at the extreme limits for survival of the fly, where both
contact. Climatic stress, lack of natural host where humans have the density of the fly is low and “personal” contact with humans may
destroyed wild animals close to villages, or clearing vegetation for be highest.
cultivation are all examples of restrict movements of palpalis group
vectors. Chemical Control

Epidemiology of East African Sleeping Sickness There are several different control techniques available
today, but the use of chemicals in controlling tsetse populations is
East African sleeping sickness differs from West African still the most common method. In brief, whether aerial or from the
sleeping sickness in both its epidemiology as well as its clinical ground, residual insecticides such as organochlorines (DDT, Dieldrin,
manifestations in mammalian host. The clinical symptoms of East Endosulfan), pyrethroids (deltamethrin, pyrethoids (deltamethrin,
African sleeping sickness are more severe, and the onset of disease is permethrin, and alphamethrin), and avermectins (ivermectin) are used
rapid. In contrast to T. b. gambiense, T.b. rhodesiense occurs with as a target areas where human-to-fly contact are likely. Pyrethroids
higher levels of parasitemia in ungulates, and humans are the are preferred because they are rapidly degraded in soil and are
adventitious hosts. The vectors of T.b. rhodesiense and the environmentally safe, unlike organochlorines, carbamates and
G.morsitans subspecies, G.pallidipes and G.swnnertoni species from organophospates that a bioaccumulate in the food chain and are
the morsitans group, and on lesser occasions the peridomestic vectors highly toxic to mammals and other vertebrates. Despite being
from the palpalis group, G.fuscipes and G.tachinoides. Sporadic cases effective, the use of organochlorines and organophosphates are now
usually arise from among those in the population whose activities banned for widespread outdoor spraying. Susceptibility to
bring them into contact with the savannah woodland habitats of the insecticides varies from one species to another, and between the
morsitans group. Although the vectors normally feed on game different classes of species.
animals, under extreme situations where “personal” contact is
increased due to social and environmental factors, a human-fly- Targets and Traps
human transmission cycle may ensue resulting in an outbreak.
Droughts and political turmoil are known to increase the number of Traps and target are mechanical devices used to kill or
cases when entire communities relocate to hitherto unoccupied areas weaken tsetse flies through insecticides or various trapping methods.
in search of safety or fertile lands and water. The use of traps and targets to control tsetse populations have been
successful primarily because tsetse flies are k-strategies with a low
rate of reproduction, and require very little sustained mortality
pressure to bring about a reduction in population or even eradication
PREVENTION AND CONTROL from an area. Hargrove estimated that an additional mortality of 4%
per day imposed on female flies was enough to cause extinction, in
Prevention the absence of immigration. The traps and targets attract tsetse flies
by taking advantage of their host-seeking behaviors, visual and
olfactory stimulation. The developments of potent attractants in the
There is no available vaccine and Chemoprophylaxis in
last 20 yrs as well as the production of second-generation synthetic
unavailable. Trypanosomes are able to switch at random to over
pyrethroid insecticides are making this form of control technique
1000, different antigens making it difficult for the body to create the
highly successful.
appropriate antibodies
Bush Clearing
Preventive measures can help:
Exploiting the knowledge that tsetse flies concentrated in
1. Wear protective clothing, including long-sleeved shirts and certain areas lead to numerous bush-clearing projects all over West
pants. The tsetse fly can bite through thin fabrics, so and East Africa to drastically alter and maintain the area unsuitable
clothing should be made of medium-weight material. for tsetse fly habitation. Discriminative bush clearing was used in
2. Wear neutral-colored clothing. The tsetse fly is attracted to Uganda to control G.m. centralis is by clearing taller Acacia trees in
bright colors and very dark colors. the Ankole district. In Tanzania, between 1923 and 1930, bush-
3. Inspect vehicles for tsetse flies before entering. The flies clearing methods were also widely employed to stop the spread of
are attracted to moving vehicles. sleeping sickness epidemic in Maswa district, where G. swynnertoni
was prevalent. Similar tactics were used in Ghana to control sleeping
4. Avoid bushes. The tsetse fly is less active during the hottest
sickness around villages were human-fly contacts were high. Despite
period of the day. It rests in bushes but will bite if the apparent success of these methods, it is widely accepted that
disturbed. bush-clearing is unsuitable as a long term control measure due to the
5. Use insect repellant. Though insect repellants have not expense and speed of reinvasion, as well as the environmental
proven effective in preventing tsetse fly bites, they are
21
damage it causes through soil erosion, decreased soil fertility, and its the Bristol-Myers Squibb Company and The Gillette Company in a
adverse effects its adverse effects on water supplies product called Vaniqa TM, a topical eflornithine HCl cream to
remove facial hair. Perhaps some of the profits generated from the
Sterile Insect Techniques sale of this form of the drug will be used to underwrite the free use of
the drug in Africa; similar to what has already happened at Merck,
who donates invermectin for the treatment of river blindness, and at
One of the modern methods of non-insecticidal control is
Pfizer Inc for their azithormycin give away program for the treatment
the Sterile Insect Technique (SIT), which was first considered as a
of trachoma.
means to control tsetse by Simpson in 1958. This technique relies on
the mating of wild females with sterile male flies. Physiologically,
female tsetse flies only required to mate once to store sperm in its SURVEILLANCE
spermathecae in sufficient quantity such that fertilization can occur
over its entire reproductive life. Mating with a sterile male would
thus result in no offspring. Sleeping sickness is one of the few communicable diseases
where systematic population screening is necessary, particularly for
gambiense sleeping sickness which has a very long almost
asympotomatic period. There are several reasons for this including
Sterilization of male Tsetse flies can be carried out by: the difficulty of diagnosis, which cannot normally be made in remote
 Irradiation primary health care facilities (4), the difficulty and high risk of
Gamma rays, Beta rays treatment for the late stage, for which special skills are required, and
 Chemo sterilization the near impossibility of vector control. Therefore, the control
Bisazir, Metepa, Tepa, Apholates, Phytosterols measure most often used for gambiense sleeping sickness is
systematic screening of the population to detect all cases, including
 Physiologic sterilization those in both the first and second stage of disease, and then curing
Pyriproxyfen, Sulphaqunizaline, Chlordimeform them. Guidelines for sleeping sickness surveillance have been
developed by WHO in collaboration with sleeping sickness endemic
countries (5).
The Future of Sleeping Sickness Control
______________________________________________________
After the publication of works such as “Silent Spring”,
public awareness of the dangers associated with insecticides are
(4) Serological detection with the CATT test (Card Agglutination
increasingly changing the way we treat our environment, and the way
Trypanosomiasis Test) is commonly used in screening. However, this
we institute environmental controls. Consequently, efforts to
test has insufficient specificity (too many false positives) to be used
introduce more environmentally friendly methods of vector control,
as a definitive diagnosis. Parasitological examinations are sufficiently
such as the use of traps without insecticides, challenges us to
specific, but are not sensitive enough unless done over a period of
understand more about the vectors that transmit the disease, as well
several successive days, because the level of parasites in the blood
as the ecological balance that we - as humans - strike with them.
oscillates rapidly. If the blood is taken during the part of the cycle
when few parasites are circulating, then a parasitological examination
We live in a world where various technical means of is likely to be negative, even though the disease is present.
control are available to address the spread of the disease. However, Appropriate treatment depends on whether or not the parasite has
sleeping sickness is a disease of the developing world, where despite passed the blood/brain barrier. A spinal tap is needed for determining
the multitude of control strategies, the issues have widely been this.
neglected and abandoned. One of the key components required to
bring about effective change is to consider the sustainability of the
(5) Trypanosomiase Humaine Africaine: Surveillance
control strategy, and to encourage local communities to take
epidemiologique et systeme d'information geographique (S.I.G),
ownership over the process, thereby empowering people to take an
Geneva, World Health Organization, 1996.
active role in an environmentally conscious solution. Increasing
knowledge through culturally sensitive education, providing technical
support, and a long-term commitment of basic resources to Surveillance Strategies
beneficiary communities is essential for large-scale tsetse control.
The five alternative surveillance strategies. These were the
Alongside efforts to reduce the spread of disease through classic mobile teams, fixed-post surveillance and the less widely used
environmental controls, there is also an urgent need to improve innovative techniques of filter paper sampling by trained community
current surveillance and diagnostic procedures. Mortality can be animal health workers, either visiting the community or based at rural
drastically reduced when cases can be diagnosed early enough to health centers.
prevent the progression of late-stage sleeping sickness. Training and
resources are desperately needed in endemic areas for proper • Fixed-post or passive surveillance, where patients
diagnostics and sero-surveillance. presenting with symptoms that are difficult to diagnose or
don't respond to treatments, say for malaria, are eventually
Perhaps the most mysterious aspect of this disease relates referred to a treatment centre and tested for a variety of
to the issue of treatment options, and the availability of drugs in disease, including trypanosomiasis, and those with the
Africa. Drug and vaccine development for diseases in developing disease are eventually diagnosed. The initial screening test
countries have always been lagging, and unfortunately, trypanocidal is performed on wet blood.
drugs are no exception. An estimated 300,000 – 500,000 people are • Filter paper sampling at rural health centers, where
currently infected and suffering from the disease with no hope for community health workers based at rural health centers
treatment. In 2000, the USFDA approved the use of eflornithine by receive some training in collecting samples on filter paper
22
and then routinely test any new patients presenting © 2007 by the Infectious Diseases Society of America. All rights
themselves at the health centre for whatever reason. reserved.
• Filter paper samplings by community health workers, 1058-4838/2007/4511-0006$15.00
who have been trained in collecting filter paper samples DOI: 10.1086/522982
and then spend 20% of their time collecting samples and CSE THEME ARTICLE MAJOR ARTICLE
following up seropositive individuals. This was based on
experience in Uganda and Côte d’Ivoire. Nifurtimox‐Eflornithine Combination Therapy for Second‐Stage
• Monovalent mobile teams, the classic surveillance teams; Trypanosoma brucei gambiense Sleeping Sickness: A Randomized
in the scenario the CATT is performed on whole blood and Clinical Trial in Congo
all the parasitological tests except for lumbar punctures are
done by the team in the field. The monovalent teams work Gerardo Priotto, 1 Serena Kasparian,1 Daniel Ngouama,2
only on trypanosomiasis. Sara Ghorashian,3 Ute Arnold,3
• Polyvalent mobile teams, which operate in the same way
as monovalent teams, except that only a third of their work
Salah Ghabri, 1 and Unni Karunakara3
consists of screening for trypanosomiasis.

Alanine aminotransferase, bilirubin, and creatinine levels were


In order to standardize the results, these were calculated for
measured by a colorimetric method (Randox) in a subgroup of
an area with a human population of 100,000 people, containing ten
patients. The reagents for alanine aminotransferase were different
rural health centers, and where 20 community animal health workers
(Human) for the last 39 patients because of logistical constraints.
were operating.
Abnormal values were defined as follows: bilirubin, >17 μmol/L,
with a >1.5‐fold increase; alanine aminotransferase, >12 IU/L (as
REFERENCES determined for 43 patients using the Randox reagents) or >32 IU/L
(for female subjects) and >42 IU/L (for male subjects, as determined
for 39 patients using the Human reagents), with a >2.5‐fold increase;
www.who.int and creatinine, >80 μmol/L (for female subjects) and >97 1Epicentre,
Paris, France; 2Programme National de Lutte contre la Trypanomose
www.cdc.gov Humaine Africaine, Ministry of Health, Republic of Congo; and
3
Médecins Sans Frontières, Amsterdam, Holland
www.new-medical.net

www.thefreedictionary.com Background. Human African trypanosomiasis caused by


Trypanosoma brucei gambiense is a fatal disease. Current treatment
www.microbiologybyte.com options for patients with second‐stage disease are either highly toxic
or impracticable in field conditions. We compared the efficacy and
www.wikipedia.org safety of the nifurtimox‐eflornithine drug combination with the
standard eflornithine regimen for the treatment of second‐stage
www.pathmicro.med disease.

www.iaea.org.at Methods. A randomized, open‐label, active‐control, phase III


clinical trial comparing 2 arms was conducted at the Sleeping
www.nhm.ac.uk Sickness Treatment Center, which was run by Médecins Sans
Frontières, in Nkayi, Bouenza Province, Republic of Congo. Patients
www.medicalecology.org were screened for inclusion and randomly assigned to receive
eflornithine alone (400 mg/kg per day given intravenously every 6 h
www.medicalhelthcare.info for 14 days) or eflornithine (400 mg/kg per day given intravenously
every 12 h for 7 days) plus nifurtimox (15 mg/kg per day given orally
www.nature.com
every 8 h for 10 days). Patients were observed for 18 months. The
study's outcomes were cure and adverse events attributable to
www.virtualcentre.org
treatment.
www.dictionary.com
Results. A total of 103 patients with second‐stage disease were
www.wrongdiagnosis.com enrolled. Cure rates were 94.1% for the eflornithine group and 96.2%
for the nifurtimox‐eflornithine group. Drug reactions were frequent in
www.healthsystem.virginia.edu both arms, and severe reactions affected 25.5% of patients in the
eflornithine group and 9.6% of those in the nifurtimox‐eflornithine
group, resulting in 2 and 1 treatment suspensions, respectively. There
was 1 death in the eflornithine arm and no deaths in the nifurtimox‐
eflornithine arm.
Journal
Conclusions. The nifurtimox‐eflornithine combination appears to
1 December 2007 be a promising first‐line therapy for second‐stage sleeping sickness.
If our findings are corroborated by ongoing findings from additional
Volume 45, Number 11 sites (a multicenter extension of this study), the new nifurtimox‐
Clinical Infectious Diseases 2007; 45:1435–1442 eflornithine combination therapy will mark a major and multifaceted
advance over current therapies.
23
Received 30 March 2007; accepted 23 May 2007; electronically combination was therefore extended to a case study of 31 patients
published 22 October 2007. that yielded similar results [19].

• (See the editorial commentary by Chappuis on pages 1443– In 2003, Médecins Sans Frontières and Epicentre, in collaboration
5) with the Ministry of Health, initiated a randomized, open‐label
clinical trial in Nkayi, Republic of Congo, to evaluate the efficacy
and toxicity of the nifurtimox‐eflornithine combination with doses
Reprints or correspondence: Dr. Gerardo Priotto, Epicentre, 8 rue
equal to those in the 2 previous studies, with additional simplification
Saint‐Sabin, 75011 Paris, France (gpriotto@epicentre.msf.org).
of the administration schedule. In 2005, the study was extended to the
Democratic Republic of Congo and Uganda, in partnership with the
Human African trypanosomiasis (HAT), or sleeping sickness, Drugs for Neglected Diseases initiative, the World Health
remains a public health challenge in sub-Saharan Africa, with an Organization Special Program for Training and Research in Tropical
estimated 50,000–70,000 new cases per year, of which 20,000 are Diseases, the Swiss Tropical Institute, and national HAT programs.
detected and reported [1]. The disease is caused by the protozoan
parasite Trypanosoma brucei gambiense, which is transmitted by the
Methods
tsetse fly (Glossina species), and it progresses from the
hemolymphatic first stage to the meningoencephalitic second stage. It
is invariably fatal without appropriate treatment. Since 1949, To facilitate external comparability, the study methodology was
melarsoprol is the most commonly used treatment for second‐stage similar to the methodologies in previous clinical trials involving
HAT. This arsenical derivative is associated with severe toxic effects second‐stage HAT [18–22].
—in particular, reactive encephalopathy, which is fatal in 10%–70%
of cases and affects 5%–10% of treated patients [2, 3]. Moreover, Participants. Study participants were identified among persons
increasing melarsoprol failure rates (up to 30%) have been reported with cases diagnosed at the treatment center or during active
in several countries [4–6]. screening. Inclusion criteria were as follows: confirmed second‐stage
infection, with trypanosomes detected in specimens of blood, lymph,
Eflornithine (diethylfluoromethylornitihine), the only new drug or CSF, with >20 leukocytes/μL in CSF specimens. Exclusion criteria
registered in 58 years for the treatment of second‐stage HAT, is a were as follows: age, <15 years; pregnancy; history of second‐stage
trypanostatic that inhibits ornithine decarboxylase, an enzyme HAT treated during the preceding 36 months; severe comorbidities
essential for cell multiplication and differentiation [7–9]. It is better likely to lead to early death during the follow‐up period; hemoglobin
tolerated than melarsoprol, and its toxic effects—mainly seizures, concentration, <5 g/dL; or inability to complete 18 months of follow‐
gastrointestinal disorders, and myelosuppression—are reversible if up for other reasons.
well managed. Its efficacy is comparable to that of melarsoprol.
However, a major disadvantage of eflornithine is the mode of Three ethics committees approved the study protocol and protocol
administration, requiring 1 slow infusion every 6 h for 14 days (56 amendments: (1) the Médecins Sans Frontières Ethical Review
infusions in total), a regimen imposed by its short half‐life of 1.5–5 h Board, (2) Comité Consultatif de Protection des Personnes dans la
[10, 11]. The difficulty in administering eflornithine in resource‐poor Recherche Biomédicale (Saint‐Germain‐en‐Laye, France), and (3)
settings explains why melarsoprol continues to be the first‐line the World Health Organization Research Ethics Review Committee
treatment. (Geneva, Switzerland). The Congolese Ministry of Health issued
authorization. All participants provided written informed consent.
Nifurtimox is an orally administered drug used in the treatment of
Chagas disease (American trypanosomiasis) at dosages of 8–20 A data safety monitoring board consisting of 4 independent experts
mg/kg per day for 90–120 days. Although it has not been approved was formed before study initiation. The data safety monitoring board
for treatment of HAT, nifurtimox is used for compassionate treatment received regular reports and issued recommendations for the study
of relapsed disease. Its toxicity, which has been poorly documented, continuation.
includes mainly neurological (headache, sleep disorders, agitation,
and confusion) and gastrointestinal (anorexia, nausea/vomiting, and
Interventions. Participants were randomized into 1 of 2 arms: the
dyspepsia) dysfunctions [12, 13], some of which increase with the
eflornithine arm or the nifurtimox‐eflornithine arm. A scientific
duration of intake [14, 15]. In the 1970s and 1980s, nifurtimox was
tested empirically in several HAT case series, and the results were committee established the dosages on the basis of published and
conflicting [14–17]. These studies, which applied different treatment unpublished evidence. The investigational arm received eflornithine
regimens and evaluation criteria, are difficult to compare. (400 mg/kg per day given intravenously every 12 h for 7 days) plus
nifurtimox (15 mg/kg per day given in oral tablets every 8 h for 10
days), and the active comparator arm received eflornithine (400
Drug combinations can potentially avert or delay the emergence of mg/kg per day given intravenously every 6 h for 14 days).
drug‐resistant organisms. Dosage reductions of each drug combined Eflornithine was infused over a 2‐h period and was diluted in 250 mL
may reduce the overall toxicity while maintaining good efficacy. of normal saline. Nifurtimox doses were repeated if vomiting
Combinations may also allow for a simpler administration of occurred within 30 min. All doses were directly administered and
treatment, improving the feasibility of therapy in remote areas with observed by the medical staff. Before commencement of treatment,
logistic and staffing limitations. all patients with malaria (as determined by microscopic evaluation
and rapid diagnostic testing) received arthemeter‐lumefantrine for 3
A first attempt to assess various combinations for the treatment of days; study treatment was started at least 1.5 days after the last dose
second‐stage HAT in Uganda was interrupted because of excess of antimalarial therapy. The administration of drugs for all other
fatality in the melarsoprol‐nifurtimox arm. Although the results were concomitant conditions was postponed until the end of the
inconclusive, the trial showed promising safety and efficacy results hospitalization, unless the clinical need warranted immediate
with the nifurtimox‐eflornithine combination [18]. Assessment of this treatment. Patients and attendants received a food ration of at least
2100 kcal/day each.
24
All patients were examined daily and hospitalized for 7 days after the that had decreased by >30%. Neutropenia as a neutrophil count
end of treatment (or longer, if deemed necessary). A lumbar puncture <2000 cells/μL that had decreased by >30%.
was performed on the day after the patient received his or her last
dose, and CSF specimens were examined for trypanosomes. μmol/L (for male subjects), with a >1.5‐fold increase.
Laboratory studies, including lumbar puncture examinations and
examinations of blood and lymph specimens, were performed at 3, 6,
12, and 18 months. At each follow‐up visit, the CSF specimen was Pharmacokinetic analysis of plasma and CSF drug concentrations
examined for parasites after double‐centrifugation, a parallel CSF was performed in an ancillary study related to the clinical trial. The
leukocyte count was determined, and IgM titers in CSF were methodology and results will be reported elsewhere.
determined using the Latex/IgM reagent (Institut de Médecine
Tropicale) [23], which aimed at increasing the sensitivity of detection Outcomes. The primary outcome was cure. The following end
of relapses. Blood samples were examined by capillary tube points were regarded as therapeutic failures: (1) death in temporal
centrifugation and quantitative buffy coat [24]. Lymph was aspirated relation to treatment (i.e., 30 days after the commencement of
from any palpable posterior cervical lymph node. treatment), and (2) relapse of HAT or death compatible with HAT
within the 18 months of follow‐up. Deaths due to disease without
The study follow‐up period was set at 18 months, as recommended clearly established alternative causality were regarded as compatible
by the Informal Consultation on the Conduct of Clinical Trials in with HAT. Secondary outcomes were the adverse events in temporal
HAT (World Health Organization, 2004) on the basis of data that relation to treatment, particularly the major adverse events graded as
suggested that 70%–90% of relapses occur 18 months after the severe (grade 3) and very severe (grade 4).
completion of treatment. Although the study end point was
determined at 18 months, the national protocol required a last follow‐ Sample size. The sample size to test non inferiority in cure rates
up visit at 24 months for all patients. for the complete multicenter study was set at 280 subjects. This
report analyzes the data of the 103 patients enrolled in Nkayi.
The definition of relapse is not currently standardized. Relapse was
diagnosed if trypanosomes were seen in body fluid specimens or if The randomization list (in blocks of 10) was electronically generated.
CSF leukocyte counts increased twice consecutively by at least 20 The list and the block size were concealed from the field team.
cells/μL any time after the completion of treatment. Patients who Participants were enrolled in the same order in which they received
presented with a single increase were reexamined 1 month later. At diagnoses. Sealed and numbered opaque envelopes containing the
the month 18 examination, relapse was diagnosed if the CSF treatment allocation were opened in strict numeric sequence.
leukocyte count was 20 cells/μL, regardless of previous counts. Blinding was unfeasible owing to the different drug administration
Distinction between relapse and reinfection was not made. The modes.
probability of reinfection was considered to be minimal, because
disease transmission had been substantially reduced after 3 years of Data management. Data were collected in purposely designed
intensive disease‐control activities by Médecins Sans Frontières. patient charts. Trial‐specific data were extracted onto case report
forms. These data were double‐entered electronically with EpiData,
Safety was assessed with the international Common Toxicity Criteria version 3.0 (The EpiData Association), and were analyzed with Stata,
[25], which grade adverse events by intensity from 1 to 4 (for mild, version 9.0 (Stata). No statistical comparisons were performed,
moderate, severe, and very severe, respectively), drug‐event because interim analysis would alter the statistical power of the
relationship (unrelated, unlikely, possible, probable, and definite), overall trial.
and outcome (complete recovery, still present, sequelae, and death).
All patients had a blood sample taken before and after treatment; the Result
sample was examined for hemoglobin concentration, total and
differential leukocyte counts, total bilirubin level, creatinine level,
and alanine aminotransferase level. Participants. Of 630 patients who had HAT diagnosed during the
trial period, 261 had cases that were in the second stage, of which
103 met the entry criteria and were enrolled in the study (figure 1).
Anemia was defined as a hemoglobin concentration <13 g/dL for The main reasons for ineligibility were relapsed status, young age,
male patients and <11 g/dL for female patients that had decreased by and/or low CSF leukocytes count. All enrolled patients were treated:
>20%. Leukopenia was defined as a leukocyte count <4000 cells/μL 51 were treated with eflornithine, and 52 were treated with
nifurtimox‐eflornithine.

25
Figure 1. Trial profile. E, eflornithine; HAT, human African trypanosomiasis; N+E, nifurtimox‐eflornithine. aRelapses were detected at 12
months in one patient and 18 months in the other. bRelapses were detected both at 18 months. cCondition was controlled at 12 months, with favorable
evolution; patient moved away later. dOne patient had controlled disease at 3 and 6 months, with favorable evolution; the patient later died of cerebral
malaria. The other patient had controlled disease at 3, 6, and 12 months, with favorable evolution; the patient later died of ovarian cancer.

Enrollment started in August 2003 and was closed in December 2004 because of a decrease in the disease prevalence in the area, resulting in a low
enrollment rate. Patient characteristics were similar in the 2 groups (table 1). Four patients (2 in each arm) had CSF leukocyte counts of 5–20
cells/μL and were wrongly enrolled. Because these 4 patients had trypanosomes in the CSF, they were kept in the study. All patients received
complete treatment in accordance with the protocol.

Table 1. Baseline characteristics of trial participants, by treatment arm.

Characteristic Eflornithine arm Nifurtimox‐


(n = 51) eflornithine arm
(n = 52)

Demographic characteristics

Female sex 23 (45.1) 26 (50.0)

Age, mean years (range) 36.1 (15–70) 33.1 (15–69)

Weight, mean kg ± SD 53.1 ± 7.2 51.7 ± 7.4

Mean body mass index a ± SD 19.7 ± 2.2 19.1 ± 2.0

Body mass index a <18.5 15 (29.4) 22 (42.3)

Parasitologic findings

Case detected by active screening 29 (56.9) 31 (59.6)

Presence of trypanosomes

In lymph nodes 40 (78.4) 36 (69.2)


26
In blood 47 (92.2) 42 (80.8)

In CSF 37 (72.6) 30 (57.7)

Leukocyte count in CSF

5–20 cells/μL 2 (3.9) 2 (3.9)

21–99 cells/μL 10 (19.6) 19 (36.5)

100 cells/μL 39 (76.5) 31 (59.6)

IgM titer in CSF >1:128 b 28 (57.1) 20 (40.8)

Clinical characteristics

Mean Karnofsky score ± SD 84.6 ± 9.6 80.9 ± 15.9

Mean Glasgow coma score ± SD 15.0 ± 0.1 14.8 ± 0.8

Hemoglobin concentration, mean g/dL ± SD 12.7 ± 1.9 13.0 ± 1.8

Presence of malaria parasites 14 (27.5) 11 (21.2)

Lymphadenopathy 40 (78.4) 36 (69.2)

Hepatomegaly 2 (3.9) 5 (9.6)

Splenomegaly 14 (27.5) 11 (21.2)

Headache 35 (68.6) 38 (73.1)

Fever c 13 (25.5) 2 (3.8)

Pruritus 33 (64.7) 33 (63.5)

Daytime somnolence 35 (68.6) 32 (61.5)

Insomnia 18 (35.3) 16 (30.8)

History of seizures 5 (9.8) 4 (7.7)

Psychiatric signs 25 (49.0) 21 (40.4)

Speech disorder 8 (15.7) 9 (17.3)

Impotence or amenorrhea 15 (29.4) 19 (36.5)

Tremors 20 (39.2) 18 (34.6)

Anorexia 6 (11.8) 12 (23.1)

Duration of symptoms, mean months (range) 9.8 (1–72) 8.7 (0–24)

Table 1. Baseline characteristics of trial participants, by treatment arm.

Outcomes and estimation. One patient died in temporal relation to the treatment regimen and was considered to have experienced treatment
failure. The large majority of patients (98 [95.1%] of 103) completed the 18‐month follow‐up period or reached the study end point earlier, and the
rest (5 [4.9%]) underwent partial follow‐up. Of the 5 patients who underwent partial follow‐up, all had demonstrated decreasing CSF leukocyte
counts and IgM titers at their last follow‐up visit. One died of cerebral malaria (last follow‐up visit was at month 6), another died after ovarian cancer
surgery (last follow‐up visit was at month 12), and 3 moved away after the month 12 follow‐up visits. There were 4 relapses in total (2 per arm). All
4 patients who experienced relapse had increasing CSF leukocyte counts without detected trypanosomes; 1 patient had relapse at 12 months, and 3

27
had relapse at 18 months. Cure rates were 94.1% (48 of 51 patients) in the eflornithine arm and 96.2% (50 of 52) in the nifurtimox‐eflornithine arm.
If patients with partial follow‐up are excluded from analysis, the cure rates are 93.3% (46 of 49 patients) in the eflornithine arm and 95.9% (45 of 49)
in the nifurtimox‐eflornithine arm.

Paired before‐after treatment CSF IgM titers were available for 88 patients. Blood contamination of CSF (6% of specimens) and other constraints
(1% of specimens) explain the missing measurements. Lumbar puncture was performed 12 h after the last dose of eflornithine and revealed that the
titers decreased in 40 (46%) of 88 patients, stayed unchanged in 38 patients (43%), and increased in 10 patients (11%). The evolution of the IgM titer
values during follow‐up ( ) showed a consistent decrease over time. Only in patients who experienced relapse did increases in IgM titers
accompany increases in CSF leukocyte counts.

Drug reactions. Only 1 patient, who was in the eflornithine group, died within 30 days after the start of treatment; this death was attributed to
septic shock following severe neutropenia. Of a total of 261 adverse events, 14 were classified as unrelated to treatment, and 247 were regarded as
drug reactions (table 2). Noteworthy differences in overall toxicity included fever, hypertension, and diarrhea, all of which were less common in the
nifurtimox‐eflornithine arm. On the other hand, the combination more frequently provoked nausea and vomiting, which tended to appear 1 h after
the simultaneous administration of both drugs (for the first daily dose) and less frequently when the drugs were administered at different times.

Table 2. Clinical and biological drug reactions during hospitalization.

Event No. (%) of events

Eflornithine arm Nifurtimox‐


(n = 51) eflornithine arm
(n = 52)

All Major All Major

Treatment‐related death 1 (2.0) … 0 (0.0) …

Neurologic reactions

Seizures 2 (3.9) 2 4 (7.7) 4

Confusion 0 (0.0) … 1 (1.9) …

Anxiety and/or agitation 4 (7.8) … 2 (3.8) …

Dizziness 1 (2.0) … 1 (1.9) …

Insomnia 1 (2.0) … 2 (3.8) …

Gastrointestinal reactions

Anorexia 0 (0.0) … 1 (1.9) …

Abdominal pain 11 (21.6) … 10 (19.2) …

Diarrhea 4 (7.8) … 0 (0.0) …

Constipation 0 (0.0) … 2 (3.8) …

Nausea and/or vomiting 8 (15.7) … 26 (50.0) …

Cardiovascular reactions

Arrythmia 3 (5.9) … 1 (1.9) …

Hypertension 8 (15.7) 1 1 (1.9) …

Edema 4 (7.8) … 0 (0.0) …

Infection

Tissue infection 1 (2.0) … 1 (1.9) …

28
Septic shock (neutropenic) 1 (2.0) 1 0 (0.0) …

Other infection 2 (3.9) 1 3 (5.8) …

Other clinical events

Fever a 13 (25.5) 2 5 (9.6) 1

Headache 15 (29.4) … 13 (25.0) …

Cough 3 (5.9) … 0 (0.0) …

Pruritus 7 (13.7) … 3 (5.8) …

Skin rash 3 (5.9) … 0 (0.0) …

Chest pain 3 (5.9) … 0 (0.0) …

Myalgia and/or arthralgia 3 (5.9) … 3 (5.8) …

Other 2 (3.9) … 4 (7.7) …

Biological reactions

Anemia 11 (21.6) 1 4 (7.7) 0

Leukopenia 1 (2.0) 0 2 (3.8) 0

Neutropenia 29 (56.9) 6 11 (21.2) 1

Abnormal bilirubin levelb 1 (2.0) … 2 (3.8) …

Abnormal alanine aminotransferase levelc 0 (0.0) … 0 (0.0) …

Abnormal creatinine levelc 0 (0.0) … 1 (1.9) …

Weight loss 5% 0 (0.0) … 3 (5.8) …

Total no. of eventsd 141 14 106 6

No. (%) of patients who experienced major events … 13 (25.5) … 5 (9.6)

Treatment interruption 2 (3.9) … 1 (1.9) …

Treatment suspension 2 (3.9) … 1 (1.9) …

Treatment termination 0 (0.0) … 0 (0.0) …

Table 2. Clinical and biological drug reactions during hospitalization.

There were 14 major drug reactions (i.e., those of grades 3 and 4) in the eflornithine arm and 6 such reactions in the nifurtimox‐eflornithine arm, and
the proportions of patients who experienced major reactions were 25.5% and 9.6% per arm, respectively (table 2). Treatment was suspended because
of severe complications for 2 patients in the eflornithine arm and 1 patient in the nifurtimox‐eflornithine arm. These interruptions were made as a
result of seizures (1 patient in each arm) and arrhythmia (1 in the eflornithine arm). The 6 major adverse events observed in the nifurtimox‐
eflornithine arm were seizures (4 patients), fever (1 patient), and neutropenia (1 patient), all of which resolved favorably.

Before‐and‐after treatment hematologic data were available for all patients. Neutropenia and anemia were the most common biological adverse
events, appearing 3 times more frequently in the eflornithine arm. Grade 3 neutropenia (i.e., a neutrophil count <1000 cells/μL) developed in 6
patients in the eflornithine arm, compared with only 1 patient in the nifurtimox‐eflornithine arm. Abnormal biochemical values were rare and mild.

29
Discussion The short half‐life of eflornithine theoretically requires shorter
administration intervals for effective therapy, and this was one of the
The results obtained with the nifurtimox‐eflornithine combination are key issues addressed in this study. We hypothesize that this adverse
similar to those obtained with standard eflornithine treatment, even pharmacokinetic profile may be balanced by the long‐lasting
showing a trend of superiority in the safety parameters. These data pharmacodynamic effect on trypanosomes, explained by the long
confirm the observations made in our earlier studies from Uganda time (18–19 h) needed by T. brucei gambiense to replenish their
[18, 19]. With nearly complete follow‐up data for patients (100% ornithine decarboxylase after inhibition by eflornithine [26]. This
reviewed at least 2 times and 95% with complete follow‐up), the high would give sufficient opportunity for trypanocydal nifurtimox to
cure rates are reassuring. eliminate the parasites. Our data seem to confirm that the 12‐h
intervals are possible if the agent is combined with nifurtimox.
The fact that 3 of the 4 relapses were detected at the month 18 visit,
having been controlled at 12 months, suggests that the follow‐up in The simplified regimen in this nifurtimox‐eflornithine schedule,
clinical studies should be no shorter than 18 months. The lower which involves 4‐fold fewer eflornithine infusions (i.e., 14 infusions
occurrence of major drug reactions in the nifurtimox‐eflornithine arm instead of 56), represents an important advance in terms of access to
suggests a better safety profile of the combination. safer and effective treatment. Most treatment centers are located near
disease transmission foci, in remote areas where logistical means and
trained staff are scarce, and most patients need treatment for second‐
Neutropenia occurred more frequently among recipients of stage disease. The twice‐per‐day infusions are key because they fit
eflornithine alone than among recipients of combination treatment, well in the routine of rural health facilities. Another important
which contains half as much eflornithine. Patients with grade 3 advantage is the cost reduction coming from the shorter
neutropenia are vulnerable to bacterial infection, which explains the hospitalization durations and from the 4‐fold reduction in the number
frequent infectious complications emerging during or after treatment, of intravenous infusions, requiring fewer infusion materials and
as well as the only death in our cohort. The combination clearly logistics and less staffing.
offers improved safety over melarsoprol, which causes reactive
encephalopathy in 5%–10% of recipients, with a high fatality rate.
There were no clinical data to support theoretical concerns about A degree of protection against the development of drug resistance can
toxicity related to drug interactions with the combination therapy. also be expected from combination versus monotherapy, as is the
case for drug combinations in use for other infectious diseases. In the
context of increasing parasite resistance to melarsoprol, and because
Nkayi presented unusual advantages in comparison to most other eflornithine is the only alternative agent for second‐stage HAT, the
HAT foci, including sociopolitical stability, the supply of electricity, availability of a combination treatment regimen is urgent, to avert the
the availability of internet communication, daily flights to the capital, development of eflornithine resistance as well.
good hospital infrastructure, and qualified medical staff. The study
did not achieve the planned sample size of 280 subjects in Nkayi
because of bureaucratic delays that resulted in the bulk of patients Because these new data reinforce the evidence in favor of the
being detected and treated by Médecins Sans Frontières before nifurtimox‐eflornithine combination, timely completion of the
enrollment could begin. There were, however, some advantages to additional sites and ensuring good follow‐up of patients are essential.
this situation: the smaller caseload facilitated good medical It will also be necessary to organize field studies with simpler—but
supervision and eased laboratory work. With a decreased prevalence sound—methodology to assess the administration of treatment in
of disease, the probability of reinfection during follow‐up was also more‐realistic situations. Studies of children will also be needed. For
minimized. all this to be possible, the continued production and availability of
nifurtimox must be ensured. If our findings are corroborated by
ongoing studies from additional sites (i.e., from a multicenter
Limitations. As a result of the insufficient number of patients extension of this study), the new nifurtimox‐eflornithine combination
recruited at this site, we have not yet performed the noninferiority therapy will mark a major and multifaceted advance over current
analysis designed in the protocol; this will only be possible when the therapies.
studies from additional sites are completed. Therefore, these data
should not be regarded as definitive proof.

Overall evidence. In the face of unsatisfactory therapeutic options


for second‐stage sleeping sickness, the need to identify new, safe, Acknowledgments
feasible, and effective therapies is urgent [3]. Research is hampered
by the fact that foci with high caseloads that can sustain good We thank the Congolese Ministry of Health—in particular, Claude
enrollment rates are often affected by conflict and located in isolated Manthelot and Damase Bodzongo, whose cooperation in setting up
and impoverished parts of Africa. Gathering the minimal elements for the study was invaluable. We acknowledge the clinical and laboratory
quality research projects (e.g., qualified personnel, infrastructure, field team members, international and local, whose hard work
communications, and stable and functional logistics) at such sites permitted this research to happen. The following individuals
represents a challenge that very few researchers are willing to contributed to the protocol development in the scientific committee:
assume. Christian Burri, Cyrus Bacchi, Dominique Legros, François
Chappuis, Marc Gastellu‐Etchegorry, Philippe Büscher, and Simon
Even if, at this first study site, we did not attain the complete sample von Nieuwenhove. The following provided laboratory technical
size, we believe that our data are of crucial interest because of the advice and training: Olivier Denis, Christian Kibonge, Barrie
promising efficacy and safety results of the nifurtimox‐eflornithine Rooney, Marina Pozzoli, Laurence Bonte, Philippe Büscher, and
combination, which was tested here for the first time with a simpler Veerle Lejon. Loretxu Pinoges kindly provided advice on statistical
(twice‐daily) eflornithine administration schedule. matters. We are indebted to the members of the Data and Safety
Monitoring Board: Dr. Pieter De Raadt, Dr. Jean Dupouy‐Camet, Dr.
Marleen Boelaert, Dr. Jacques Chandenier and Joris Menten. The

30
Drugs for Neglected Diseases initiative and the Swiss Tropical • 8. Bacchi CJ, Nathan HC, Hutner SH, McCann PP,
Institute provided advice on the Good Clinical Practice guidelines. Sjoerdsma A. Polyamine metabolism: a potential
therapeutic target in trypanosomes. Science 1980; 210:332–
Financial support. This study was funded by the Dutch section of 4.
Médecins Sans Frontières. The Médecins Sans Frontières
International Nobel Prize Fund supported the original protocol First citation in article, CrossRef, PubMed
development.
• 9. Bacchi CJ, Garofalo J, Mockenhaupt D, et al. In vivo
Potential conflicts of interest. All authors: no conflicts. effects of alpha‐DL‐difluoromethylornithine on the
metabolism and morphology of Trypanosoma brucei
References brucei. Mol Biochem Parasitol 1983; 7:209–25.

• 1. World Health Organization. Human African First citation in article, CrossRef, PubMed
trypanosomiasis (sleeping sickness): epidemiological
update. Wkly Epidemiol Rec 2006; 81:71–80. Available at: • 10. Abeloff MD, Slavik M, Luk GD, et al. Phase I trial
http://www.who.int/wer/2006/wer8108.pdf. and pharmacokinetic studies of alpha‐
difluoromethylornithine—an inhibitor of polyamine
First citation in article biosynthesis. J Clin Oncol 1984; 2:124–30.

• 2. Pepin J, Milord F, Khonde AN, et al. Risk factors for First citation in article, PubMed
encephalopathy and mortality during melarsoprol treatment
of Trypanosoma brucei gambiense sleeping sickness. Trans • 11. Griffin CA, Slavik M, Chien SC, et al. Phase I trial
R Soc Trop Med Hyg 1995; 89:92–7. and pharmacokinetic study of intravenous and oral alpha‐
difluoromethylornithine. Invest New Drugs 1987; 5:177–
First citation in article, CrossRef, PubMed 86.

• 3. World Health Organization. Control and surveillance of First citation in article, CrossRef, PubMed
African trypanosomiasis: report of a WHO expert
committee. WHO technical report series 881. Geneva: • 12. Wegner DH, Rohwedder RW. The effect of nifurtimox
World Health Organization, 1998. in acute Chagas’ infection. Arzneimittelforschung 1972;
22:1624–35.
First citation in article

• 4. Burri C, Keiser J. Pharmacokinetic investigations in


patients from northern Angola refractory to melarsoprol First citation in article, PubMed
treatment. Trop Med Int Health 2001; 6:412–20.

• 13. Wegner DH, Rohwedder RW. Experience with


First citation in article, CrossRef, PubMed nifurtimox in chronic Chagas’ infection: preliminary report.
Arzneimittelforschung 1972; 22:1635–41.
• 5. Legros D, Evans S, Maiso F, Enyaru JC, Mbulamberi
D. Risk factors for treatment failure after melarsoprol for First citation in article, PubMed
Trypanosoma brucei gambiense trypanosomiasis in
Uganda. Trans R Soc Trop Med Hyg 1999; 93:439–42.
• 14. Janssens PG, De Muynck A. Clinical trials with
“nifurtimox” in African trypanosomiasis. Ann Soc Belg
First citation in article, CrossRef, PubMed Med Trop 1977; 57:475–80.

• 6. Stanghellini A, Josenando T. The situation of sleeping First citation in article, PubMed


sickness in Angola: a calamity. Trop Med Int Health 2001;
6:330–4.
• 15. Pepin J, Milord F, Meurice F, Ethier L, Loko L, Mpia
B. High‐dose nifurtimox for arseno‐resistant Trypanosoma
First citation in article, CrossRef, PubMed brucei gambiense sleeping sickness: an open trial in central
Zaire. Trans R Soc Trop Med Hyg 1992; 86:254–6.
• 7. Metcalf BW, Bey P, Danzin C, Jung MJ, Casara P,
Vevert JP. Catalytic irreversible inhibition of mammalian First citation in article, CrossRef, PubMed
ornithine decarboxylase (E.C.4.1.1.17) by substrate and
product analogues. J Am Chem Soc 1978; 100:2551–3.
• 16. Moens F, De Wilde M, Ngato K. [Clinical trial of
nifurtimox in human African trypanosomiasis]. Ann Soc
First citation in article, CrossRef Belg Med Trop 1984; 64:37–43.

31
First citation in article, PubMed • 25. National Cancer Institute, National Institutes of
Health. Cancer therapy evaluation program: common
• 17. Van Nieuwenhove S. Advances in sleeping sickness toxicity criteria, version 2.0. 1999. Available at:
therapy. Ann Soc Belg Med Trop 1992; 72(Suppl 1):39–51. http://ctep.cancer.gov/forms. Accessed 21 May 2007

First citation in article, PubMed First citation in article

• 18. Priotto G, Fogg C, Balasegaram M, et al. Three drug • 26. Iten M, Mett H, Evans A, Enyaru JC, Brun R,
combinations for late‐stage Trypanosoma brucei Kaminsky R. Alterations in ornithine decarboxylase
gambiense sleeping sickness: a randomized clinical trial in characteristics account for tolerance of Trypanosoma
Uganda. PLoS Clin Trials 2006; 1:e39. brucei rhodesiense to D,L‐alpha‐difluoromethylornithine.
Antimicrob Agents Chemother 1997; 41:1922–5.
First citation in article
First citation in article, PubMed

• 19. Checchi F, Piola P, Ayikoru H, Thomas F, Legros D,


CSE Global Theme Issue on Poverty and Human Development
Priotto G. Nifurtimox plus eflornithine for late‐stage
sleeping sickness in Uganda: a case series. PLoS Neglected
Trop Dis (in press). Cited by

First citation in article D. Steverding, X. Wang. (2009) Evaluation of anti-sleeping-sickness


drugs and topoisomerase inhibitors in combination on Trypanosoma
brucei. Journal of Antimicrobial Chemotherapy 63:6, 1293-1295
• 20. Burri C, Nkunku S, Merolle A, Smith T, Blum J, Brun
R. Efficacy of new, concise schedule for melarsoprol in
Online publication date: 1-Jul-2009.
treatment of sleeping sickness caused by Trypanosoma
CrossRef
brucei gambiense: a randomised trial. Lancet 2000;
D. Mumba Ngoyi, V. Lejon, F.X. N’Siesi, M. Boelaert, P.
355:1419–25.
Büscher. (2009) Comparison of operational criteria for treatment
outcome I n gambiense human African trypanosomiasis. Tropical
First citation in article, CrossRef, PubMed Medicine & International Health 14:4, 438-444
Online publication date: 1-May-2009.
• 21. Pepin J, Khonde N, Maiso F, et al. Short‐course CrossRef
eflornithine in Gambian trypanosomiasis: a multicentre Peter G. E. Kennedy. (2008) The continuing problem of human
randomized controlled trial. Bull World Health Organ African trypanosomiasis (sleeping sickness). Annals of Neurology
2000; 78:1284–95. 64:2, 116-126
Online publication date: 1-Sep-2008.
CrossRef
First citation in article, PubMed Veerle Lejon, Isabelle Roger, Dieudonné Mumba Ngoyi,
Joris Menten, Jo Robays, Francois X. N’Siesi, Sylvie Bisser,
• 22. Schmid C, Nkunku S, Merolle A, Vounatsou P, Burri Marleen Boelaert, and Philippe Büscher. (2008) Novel Markers for
C. Efficacy of 10‐day melarsoprol schedule 2 years after Treatment Outcome in Late‐Stage Trypanosoma brucei gambiense
treatment for late‐stage gambiense sleeping sickness. Trypanosomiasis. Clinical Infectious Diseases 47:1, 15-22
Lancet 2004; 364:789–90. Online publication date: 1-Jul-2008.
Abstract-Full Text-PDF Version (402 kB)
François Chappuis
First citation in article, CrossRef, PubMed CSE Poverty and Human Development. (2007) Editorial
Commentary: Melarsoprol‐Free Drug Combinations for Second‐
• 23. Lejon V, Buscher P, Sema NH, Magnus E, Van Stage Gambian Sleeping Sickness: The Way to Go. Clinical
Meirvenne N. Human African trypanosomiasis: a latex Infectious Diseases 45:11, 1443-1445
agglutination field test for quantifying IgM in cerebrospinal Online publication date: 1-Dec-2007.
fluid. Bull World Health Organ 1998; 76:553–8. Citation-Full Text-PDF Version (162 kB)

First citation in article, PubMed

Medical Research
• 24. Bailey JW, Smith DH. The use of the acridine orange
QBC technique in the diagnosis of African
trypanosomiasis. Trans R Soc Trop Med Hyg 1992; 86:630. Submitted to: Dr.Mazhar Qayyum
Submitted by: Farthat Yasmeen
First citation in article, CrossRef, PubMed Date: 23/o6/2008

Trypanosoma brucei

32
Scientific Classification • T. brucei gambiense - Causes slow onset chronic
trypanosomiasis in humans. Most common in central and
Kingdom: Protista western Africa, where humans are thought to be the
primary reservoir.
Phylum: Euglenozoa
• T. brucei rhodesiense - Causes fast onset acute
trypanosomiasis in humans. Most common in southern and
Subphylum: Mastigophora
eastern Africa, where game animals and livestock are
thought to be the primary reservoir.
Class: Kinetoplastoa
• T. brucei brucei - Causes animal African trypanosomiasis,
Order: Trypanosomastida along with several other species of trypanosoma. T. b.
brucei is not human infective due to its susceptibility to
Genus: Trypanosoma lysis by human apolipoprotein L1. However, as it shares
many features with T. b. gambiense and T. b. rhodesiense
Species: Trypanosoma brucei (such as antigenic variation) it is used as a model for
human infections in laboratory and animal studies.

The cell structure


Description and Significance:
The structure of the cell is typical of eukaryotes, see
eukaryotic cell. All major organelles are seen, including the nucleus,
The genus Trypanosoma contains a large number of mitochondria, endoplasmic reticulum, Golgi apparatus etc. Unusual
parasitic species, which infect wild and domesticated animals and features include the single large mitochondria with a condensed
humans in Africa. Commonly known as African sleeping sickness, mitochondrial DNA structure, and its association with the basal body
human trypanosomiasis is caused by the species Trypanosoma brucei of the flagellum, unusually the cytoskeleton organization mechanism
and is transmitted to human through either a vector or the blood of of the cell. The cell surface of the bloodstream form features a dense
ingested animal. The most common vector of Trypanosoma brucei is coat of variable surface glycoproteins (VSGs) which is replaced by
the tsetse fly, which may spread the parasite to humans and animals an equally dense coat of procyclins when the parasite differentiates
through bites. Through the process of antigenic variation, some into the procylic in the tsetse fly midgut. Trypanosomatid cellular
trypanosomes are able to evade the host’s immune system by forms.
modifying their surface membrane, essentially multiplying with every
surface change. As the disease progress, Trypanosoma brucei Trypanosomatids show specific cellular forms:
gradually infiltrates the host’s central nervous system. Symptoms
include headache, weakness, and joint pain in the initial stages;
anemia, cardiovascular problems, and kidney disorders as the disease
• Amastigote - Basal body anterior of nucleus, with a short,
essentially non-functional, flagellum.
progresses; in its final stages, the disease may lead to extreme
exhaustion and fatigue during the day, insomnia at night, coma, and • Promastigote - Basal body anterior of nucleus, with a long
ultimately death. Human trypanosomiasis affects as many as 66 detached flagellum.
million people in Sub-Saharan Africa. Trypanosomes are also found • Epimastigote - Basal body anterior of nucleus, with a long
in the America in the form of Trypanosoma cruzi, which causes flagellum attached along the cell body.
American human trypanosomiasis, or Chagas’ disease. This disease is • Trypomastigote - Basal body posterior of nucleus, with a
found in two forms: as an amastigote in the cells, and as a long flagellum attached along the cell body.
trymastigote in the blood. The vectors for Trypanosome cruzi include
members of the order Hemiptera, such as assassin flies, whichingest
These names are derived from the Greek mastig- meaning whip,
the amastigote or trymastigote and carry them to animals or humans.
referring to the trypanosome's whip-like flagellum.
The parasites enter the human host through mucus membranes in the
nose, eye, or mouth upon release from the insect vectors. Left
untreated, Chagas’ disease may cause dementia, megacolon and T. brucei is found as a trypomastigote in the slender, stumpy,
megaesophagus and damage to the heart muscle and may result in procyclic and metacyclic forms. The procylic form differentiates to
death. the proliferitive epimastigote form in the salivary glands of the insect.
Unlike Leishmania, the promastigote and the amastigote form does
The infection: Trypanosomiasis not form part of the T.brucei life cycle.

The insect vector for T. brucei is the tsetse fly. The parasite The genome
lives in the midgut of the fly (procyclic form), whereupon it migrates
to the salivary glands for injection to the mammalian host on biting. The genome of T. brucei is made up of:
The parasite lives within the bloodstream (bloodstream form) where
it can reinfect the fly vector after biting. Later during a T. brucei
infection the parasite may migrate to other areas of the host. A T. • 11 pairs of large chromosomes of 1 to 6 megabase pairs.
brucei infection may be transferred human to human via bodily fluid • 3-5 intermediate chromosomes of 200 to 500 kilobase
exchange, primarily blood transfer. pairs.
• Around 100 mini chromosomes of around 50 to 100
There are three different sub-species of T. brucei, which cause kilobase pairs. These may be present in multiple copies per
different variants of trypanosomiasis. haploid genome.

33
The large chromosomes contain most genes, while the small Antigenic variation
chromosomes tend to carry genes involved in antigenic variation,
including the VSG genes. The genome has been sequenced and is Sequencing of the T. brucei genome has revealed a huge
available online. VSG gene archive, made up of thousands of different VSG genes. All
but one of these is 'silent' VSGs, as each trypanosome expresses only
The mitochondrial genome is found condensed into the kinetoplast, one VSG gene at a time. VSG is highly immunogenic, and an
an unusual feature unique to the kinetoplastea class. It and the basal immune response raised against a specific VSG will rapidly kill
body of the flagellum are strongly associated via a cytoskeleton trypanosomes expressing this VSG. This can also be observed in
structure. vitro by a complement-mediated lysis assay. However, with each cell
division there is a possibility that one or both of the progeny will
The cytoskeleton switch expression to a silent VSG from the archive (see below). The
frequency of such a switch has been measured to be approximately
1:100. This new VSG will likely not be recognised by the specific
The cytoskeleton is predominantly made up of immune responses raised against previously expressed VSGs. It takes
microtubules, forming a subpellicular corset. The microtubules lie several days for an immune response against a specific to develop,
parallel to each other along the long axis of the cell, with the number giving trypanosomes, which have undergone VSG coat switching
of microtubules at any point roughly proportional to the some time to reproduce (and undergo further VSG coat switching
circumference of the cell at that point. As the cell grows (including events) unhindered. Repetition of this process prevents extinction of
for mitosis) additional microtubules grow between the existing the infecting trypanosome population, allowing chronic persistence of
tubules, leading to semiconservative inheritance of the cytoskeleton. parasites in the host. The clinical effect of this cycle is successive
The microtubules are orientated + at the posterior and - at the 'waves' of parasitaemia (trypanosomes in the blood).
anterior. Microfilament and intermediate filaments also play an
important role in the cytoskeleton, but these are generally
overlooked. Trypanosome cell cycle

Flagellar structure The mitotic division of T.brucei is unusual in terms of the


cytoskeletal process. The basal body, unlike a centrosome of most
eukaryotic cells, plays an important role in the organisation of the
The trypanosome flagellum has two main structures. It is spindle.
made up of a typical flagellar axoneme, which lies parallel to the
paraflagellar rod, a lattice structure of proteins unique to the
kinetoplastida, euglenoids and dinoflagellates. Stages of mitosis:

The microtubules of the flagellar axoneme lie in the normal 9+2 6. The basal body replicates, both remaining associated with
arrangement, orientated with the + at the anterior end and the - in the the kinetoplast.
basal body. The cytoskeletal structure extends from the basal body to 7. The kinetoplast undergoes replication, and the daughter
the kinetoplast. The flagellum is bound to the cytoskeleton of the kinetoplasts are separated by the basal bodies.
main cell body by four specialised microtubules, which run parallel 8. The second flagellum grows while the nucleus undergoes
and in the same direction to the flagellar tubulin. replication.
9. The mitochondrion divides, and cytokinesis progresses
from the anterior to posterior end.
The flagellar function is twofold - locomotion via oscilations along
10. The division resolves. The daughter cells may stay
the attached flagellum and cell body, and attachment to the fly gut
connected for a significant length of time after cytokinesis
during the procyclic phase.
is complete.

The VSG coat


Prevention and Control

The surface of the trypanosome is covered by a dense coat of


Infection by trypanosome species is acquired from the bite
~1x107 molecules of Variable Surface Glycoprotein (VSG).[4] This
of an infected tsetse fly. Thus, preventing flies from biting with
coat enables an infecting T. brucei population to persistently evade
repellants or insect nets will reduce the transmission of the parasite.
the host's immune system, allowing chronic infection. The two
Control of the flies through insecticides and habitat alteration
properties of the VSG coat that allow immune evasion are:
(removal of plant cover near the water sources) is possible, but has
shown to be very difficult.
• Shielding - the dense nature of the VSG coat prevents the
immune system of the mammalian host from accessing the Case management and Treatment
plasma membrane or any other invariant surface epitopes
(such as ion channels, transporters, receptors etc.) of the
parasite. The coat is uniform, made up of millions of copies There are three stages to case management:
of the same molecule; therefore the only parts of the
trypanosome the immune system can 'see' are the N- • Screening is the initial sorting of people who might be
terminal loops of the VSG that make up the coat. infected. This involves checking for clinical signs or the
• Periodic antigenic variation - the VSG coat undergoes use of serological test.
frequent stochastic genetic modification - 'switching' - • Diagnosis shows whether the parasite is present. The only
allowing variants expressing a new VSG coat to escape the sign, one that has been known for centuries, is swollen
specific immune response raised against the previous coat. cervical glands (Winterbottom sign)

34
• Phase diagnosis shows the state of progression of the The genome of the parasite has been decoded and several
disease. It entail examination of CSF obtained by lumbar proteins have been identified as potential targets for drug treatment.
puncture and is used to determine the course of treatment The decoded DNA also revealed the reason why generating a vaccine
for this disease has been so difficult. Trypanosoma brucei has over
800 genes that manufacture proteins that the disease mixes and
The long, asymptomatic first phase of T.b. gambiense sleeping
matches to evade immune system detection.
sickness is one of the factors that make treatment difficult. Diagnosis
must be made as early as possible in order to preclude the onset of
irreversible neurological disorders and prevent transmission. Case Recent findings indicate that the parasite is unable to
detection is difficult and requires major human, technical and survive in the bloodstream without its flagellum. This insight gives
material sources. Since the disease is rife in rural areas among poor researchers a new angle with which to attack the parasite.
people with little access to health facilities, this problem is all the
more difficult. A new treatment based on a truncated version of
apolipoprotein L-1 of high-density lipoprotein and a nanobody has
Treatment recently been found to work in mice, but has not been tested to
human.
If the disease is diagnosed early, the chances of cure are
high. The type of treatment depends on the phase of disease; initial or New Scientist, 25 Aug.2007, pp. 35.7 ref
neurological. Success in the latter phase depends on having a drug
that can cross the blood-brain barrier to reach the parasite. Four drug The cover story of the August 25,2006 issue of Cell journal
have been used until now. describe an advance; Dr. Lee Soo Hee and colleagues, working at
John Hopkins, have investigated the pathway by which the organism
• Suramine make myristate, a 14-carbon length fatty acid. Myristate is a
• Pentamidine component of the VSG, the molecule that makes trypanosome’s outer
• Melarsoprol layer. This outer surface coat of VSG is vital to the trypanosome’s
• Eflornithine avoidance of immunological capture, Dr. Lee and Colleague
discovered trypanosomes use a novel fatty acid synthesis pathway
involving fatty acid elongases to make myristate and other fatty
Drug Target and Drug Discovery acids.

35
36

Você também pode gostar