You are on page 1of 10

Eur J Clin Microbiol Infect Dis (2012) 31:109–118

DOI 10.1007/s10096-011-1276-0

REVIEW

Leishmaniasis: new insights from an old


and neglected disease
S. Antinori & L. Schifanella & M. Corbellino

Received: 11 February 2011 / Accepted: 12 April 2011 / Published online: 1 May 2011
# Springer-Verlag 2011

Abstract Leishmaniases are a clinically heterogeneous disease manifestations: visceral, cutaneous, and mucocuta-
group of diseases caused by protozoa of the genus neous. Despite the fact that leishmaniasis is considered to
Leishmania. There is growing evidence that the true be a neglected disease, a large body of novel findings have
incidence of the disease is underestimated, especially in been generated in the last few years on the taxonomy,
hyperendemic regions. Moreover, climate changes together epidemiology, clinical manifestations, diagnosis, and ther-
with the increasing movement of humans and animals raise apy of these infections. An analysis of the published
concerns about the possible introduction of Leishmania research on leishmaniasis over a period of 50 years
infection in previously spared areas. The disease is (1957–2007) showed an increasing number of publications
emerging in immunocompromised patients undergoing over time, which peaked in 2006 with 1,226 published
bone marrow or solid organ transplantation or treatment items [1]. The articles originated from 140 countries, with
with biologic drugs. Furthermore, the deployment of the United States of America (USA) and Brazil being the
military troops and travel to endemic areas are associated two most productive, followed by six European countries
with the observation of a growing number of patients with (i.e., United Kingdom, France, Germany, Spain, Switzer-
cutaneous disease. Improvement in diagnostic methods, land, and Italy). These same countries, with the inclusion of
both in the field and in specialized laboratories, has been Canada and India, appear in the top ten list of the most
obtained through the implementation of molecular amplifi- cited papers on the leishmaniases, with the USA and UK
cation methods and using the rK39 antigen as the substrate. ranking, respectively, first and second. Less than 20% of
Finally, new therapeutic approaches are gaining attention, the articles are the result of an international cooperation,
such as the use of miltefosine for cutaneous leishmaniasis with USA and Brazil showing the highest degree of
and paromomycin for visceral leishmaniasis, as well as the cooperation.
use of various antileishmanial drugs in combination. In the present review, we will present an update of the
most recent advances on Leishmania infections.

Introduction
Taxonomy
Leishmaniases are protozoan parasitic infections transmit-
ted to mammals (including human beings) by phlebotomine Unlike the Plasmodia, the different species of Leishmania
sandflies and they are associated with three main types of present identical morphology when classical diagnostic
methods such as microscopic examination or parasite
S. Antinori (*) : L. Schifanella : M. Corbellino culture are used. Thus, the classification of Leishmania
Department of Clinical Sciences L. Sacco, was historically based on criteria such as geographical
Section of Infectious Diseases and Immunopathology, distribution, vector, tropism, and clinical manifestation [2,
Università degli Studi di Milano,
Via GB Grassi, 74,
3]. The two known subgenera (L) Leishmania and (L)
20157 Milano, Italy Viannia were separated according to their ability to develop
e-mail: spinello.antinori@unimi.it in the foregut (Suprapylaria) and hind-midgut (Peripylaria)
110 Eur J Clin Microbiol Infect Dis (2012) 31:109–118

of the sandfly, respectively. Recognition of the species al. have been proposed as “work in progress” in the
within the subgenera was accomplished by using as the revision and simplification of Leishmania’s nomenclature,
gold standard multilocus enzyme electrophoresis (MLEE) given the caveats arising from the use of a single genetic
[4]. However, the validity of the current classification marker of the parasite [8].
system that recognizes 30 species, of which 20 are
responsible for human disease, has been repeatedly ques-
tioned [5]. Although several molecular methods have been Epidemiology
used for defining Leishmania taxonomy, this is still a matter
of increasing debate [6]. Very recently, Fraga et al. used There is a growing body of data highlighting the notion that
sequences of the highly conserved 70-kDa heat shock the global health impact of the leishmaniases is grossly
protein (hsp70 gene) to analyze isolates and strains of underestimated due to multiple factors (i.e., increasing
different geographic origin, showing that only eight prevalence of the disease, unrecorded cases, expanding
monophyletic groups were detectable against the 17 areas of endemicity). In this regard, secondary data may be
examined (Table 1) [7]. According to these authors, in the useful to calculate the true burden of the disease [9]. The
L. donovani complex, only L. donovani retained the status need to arrange reliable epidemiological surveillance data is
of species and only L. donovani infantum should be strongly supported by the final assessment of the World
considered a subspecies. Similarly, only L. tropica and not Health Organization (WHO) Elimination Initiative. It is
L. aethiopica conserved the status of species, while they well known that more than 90% of visceral leishmaniasis
proposed to consider L. major as an independent species (VL) are reported from the Indian subcontinent and Sudan,
[7]. As far as the New World Leishmania and Viannia are and that the Indian state of Bihar alone accounts for nearly
concerned, only L. (L) mexicana, L. (V) braziliensis, L. 90% of all cases. However, the 200,000 cases of L.
guyanensis, and L. (V) lainsoni were recognized as donovani infection officially reported from Bihar since
individual species, with the status of L. naiffi awaiting 1977 are, most probably, an underestimate of the real
further investigations. The results of the study by Fraga et incidence of the disease. In fact, two population-based

Table 1 Newly proposed nomenclature for Leishmania species based on the heat shock protein 70 (hsp70) gene sequence

Genus Complex Species Geographic distribution Species according


to hsp70 analysis

L. (Leishmania) L. donovani L. donovani China, Indian subcontinent, Ethiopia, Sudan, Kenya, Iran, L. donovani
Saudi Arabia, Yemen
L. infantum Albania, Algeria, France, Greece, Italy, Morocco,
Portugal, Spain, Syria, Tunisia, Turkey, Yemen
L. chagasi Argentina, Bolivia, Brazil, Colombia, Ecuador, El Salvador,
Guadalupe, Guatemala, Honduras, Martinique, Mexico,
Nicaragua, Paraguay, Suriname, Venezuela
L. archibaldi India, Sudan, Ethiopia, Lebanon, Israel
L. tropica L. tropica Afghanistan, Algeria, Azerbaijan, Greece, Iran, Iraq, L. tropica
Israel, Morocco, Tunisia, Turkey, Yemen
L. aethiopica Ethiopia, Kenya
L. major Afghanistan, Algeria, Chad, Iran, Iraq, Israel, Libya, L. major
Mauritania, Morocco, Syria, Sudan
L. mexicana L. mexicana Belize, Colombia, Costa Rica, Dominican Republic, L. mexicana
Ecuador, Guatemala, Honduras, Mexico, Panama, Venezuela
L. amazonensis Bolivia, Brazil, Colombia, Costa Rica, Ecuador, French Guyana,
Panama, Peru, Venezuela
L. garnhami Venezuela
L. (Viannia) L. guyanensis L. guyanensis Brazil, Colombia, Ecuador, French Guyana, Peru, Suriname, L. guyanensis
Venezuela
L. panamensis Belize, Colombia, Costa Rica, Ecuador, Honduras, Nicaragua,
Panama, Venezuela
L. naiffi Brazil, French Guyana, Ecuador, Peru L. naiffi
L. braziliensis L. braziliensis Argentina, Belize, Bolivia, Brazil, Colombia, Costa Rica, L. braziliensis
Ecuador, Guatemala, Honduras, Nicaragua
L. peruviana Peru
L. lainsoni Brazil, Bolivia, Peru L. lainsoni?
Eur J Clin Microbiol Infect Dis (2012) 31:109–118 111

studies conducted in Bihar’s sub-districts documented an Hosts


annual VL incidence rate of 2.5 and 5.7 cases per 1,000,
thus, supporting the fact that official figures underestimate The global spread of human immunodeficiency virus (HIV)
the incidence of the disease by a factor of 8 and 4, infection was responsible, starting from the mid-1980s, for
respectively [10, 11] . Another recent study that included in the upsurge of HIV/VL coinfection in four Mediterranean
the sample more than 5% of the population of a rural countries (i.e., Spain, France, Italy, and Portugal), with such
district of Bihar and 35% of the population of high cases occurring almost exclusively among intravenous drug
incidence areas demonstrated an annual incidence of VL users [23]. It was shown that the risk of developing overt
of 2.2 cases per 1,000 [12]; in this regard, the authors VL was increased by a factor of 100–1,000 in HIV-infected,
suggest that, in order to achieve the VL elimination goal by as opposed to HIV-uninfected, individuals [24]. HIV
2015 (i.e., less than 0.1 cases per 1,000), a 22- to 35-fold infection was also responsible in these countries for a shift
reduction of the disease’s incidence will be required. of leishmaniases from a predominantly pediatric to an adult
In Europe, both cutaneous leishmaniasis (CL) and VL disease. The peak incidence of HIV/VL coinfection was
are well established diseases in the Mediterranean basin, registered during the period from January 1996 to June
with an incidence ranging from 0.02/100,000 to 0.49/ 1998, when the introduction of highly active antiretroviral
100,000 that roughly translates to 700 cases per year therapy (HAART) became widespread in Europe [25]. In
[13]. However, there is concern that changes in the the ensuing years, a sharp decrease of HIV/VL incidence
environment, human behavior, and the climate, as well as was observed throughout southern Europe, with the
increasing dog travel, will trigger the introduction of L. possible exception of Portugal [26, 27]. On the contrary,
infantum into Northern Europe [14]. Several entomolog- HIV/VL coinfection is presently expanding in sub-Saharan
ical surveys have shown that two VL vectors (namely, Africa, the Indian subcontinent, and South America, not
Phlebotomus perniciosus and P. neglectus) have increased only as a consequence of social phenomena such as mass
in density and have expanded their range to cover the migration and wars, but also of growing local resistance to
Italian Alpine regions, where they were previously antimonials and restricted access to combination antiretro-
undetected [15, 16]. Moreover, confirmed or suspected viral therapy. For instance, in Ethiopia, 35% of all cases of
autochthonous cases of canine and human visceral VL in Addis Ababa are associated with HIV infection and
leishmaniases have been reported not only from northern higher figures are observed in the Humera area of western
Italy, but as north as Germany [17–19]. Tigray (i.e., the main region of Leishmania endemicity in
In the Eastern Mediterranean Region of the WHO, both that country) among migrant workers involved in the cotton
CL (zoonotic and anthroponotic) and VL (zoonotic) occur and sesame field agriculture, where the proportion of
in 14 of the 22 countries (i.e., Afghanistan, Egypt, Islamic patients with VL coinfected was as high as 40% in 2006
Republic of Iran, Iraq, Jordan, Libyan Arab Jamahiriya, [28]. Preliminary data obtained by Médecins Sans Fron-
Morocco, Pakistan, Saudi Arabia, Somalia, Sudan, Syrian tières in Nasir (Sudan) showed a 25% prevalence rate of
Arab Republic, Tunisia, and Yemen) [20]. In 2008, nearly HIV/VL coinfection, whereas in Khartoum, hospital-based
100,000 new cases of CL were reported from 12 countries, studies documented a prevalence of 5–9.4% [29]. In Brazil,
with the highest incidences from Afghanistan (24,585 2% of cases of VL reported in the period 2001–2005 were
cases), Syrian Arab Republic (29,140 cases), and Iran coinfected with HIV, but it is worth noting that, contrary to
(26,824 cases). However, a recent survey conducted in the what is observed in southern Europe, VL represents only
Jordan River valley found an incidence rate for CL of 75/ 37% of all clinical forms of the disease among coinfected
100,000, which is a figure higher than that observed in the cases, with mucocutaneous leishmaniases being seen in at
Syrian Arab Republic [21]. In the same year, around 5,000 least 43% of HIV-positive patients [23].
cases of VL were reported from Sudan and Somalia, with However, also in Europe, the hosts at risk of developing
another 500 cases from five countries (Iran, Morocco, VL are quite variable, as demonstrated by a recent study
Saudi Arabia, Syria, and Tunisia), but these figures conducted in Albania, where it was shown that VL is still a
probably still represent an underreporting of the total pediatric disease, with nearly 90% of the total cases still
burden of the disease [20]. observed in patients aged below 15 years, with the highest
In Latin America, Brazil is the country with the highest incidence (25/100,000) in the age group between 0 and
burden of VL, with an increased incidence from an average 6 years [30].
of 1,500 cases per year in the 1980s to an average of more The leishmaniases, both cutaneous and visceral, are, not
than 3,000 cases per year between 2000 and 2006 [22]. In unexpectedly, emerging as bona fide opportunistic infec-
this country, the disease spreading southward and eastward tions among immunocompromised hosts such as those
has been observed in the states of Saõ Paulo and Mato undergoing solid organ and bone marrow transplantation, in
Grosso do Sul. patients with autoimmune diseases being treated with
112 Eur J Clin Microbiol Infect Dis (2012) 31:109–118

immunosuppressive agents, and, more recently, with bio-


logic drugs (i.e., infliximab, etanercept, adalimumab) [31–
37]. Another category of patients who may probably be at
higher risk of developing leishmaniasis is represented by
those affected by cancer. In a recent review involving 44
patients, four possible associations between cancer and
leishmaniasis were identified: leishmaniasis mimicking
cancer; a difficult to diagnose infection in patients receiving
chemotherapy for the underlying malignancy; coexistence
of the two diseases; and, finally, the putative role of
Leishmania spp. infection as a trigger for cutaneous and
mucosal malignancies [38].
Another group of individuals who are recognized as
being at increasing risk to develop leishmaniasis are
international travellers and military personnel (Fig. 1)
deployed in areas of high endemicity [39–43]. In 2004, Fig. 2 Cutaneous leishmaniasis with ulcerated “volcano-like” appear-
ance due to L. panamensis in an Italian traveller who had visited
more than 600 cases of CL were diagnosed in American several South American countries (Costa Rica, El Salvador, Guate-
soldiers deployed to Iraq, Kuwait, and Afghanistan [42] . mala, Mexico, Panama)
Subsequently, van Thiel et al. reported an attack rate of
18.3% for CL among Dutch troops in Afghanistan [43].
In the GeoSentinel Network, CL ranked 10th among all (i.e., fever, weight loss, hepatosplenomegaly, and pancyto-
dermatologic conditions observed in returning travellers penia) are observed in about 80–90% of patients. However,
(Fig. 2); the disease was generally associated with male what is markedly different in HIV-positive patients is the
predominance and long duration of travel, as well as lower rate of response to treatment and the high rate of
Central and South American destinations (particularly disease relapse that approaches 90% in those who are not
Bolivia) [44]. receiving HAART [46]. Although HAART is associated
with a reduced incidence of VL relapse, it is not universally
effective in preventing disease recurrence. In contrast to the
Clinical aspects classic acquired immune deficiency syndrome (AIDS)-
related opportunistic infections, in the case of HIV/VL, a
Clinical manifestations of leishmaniasis depend on both the more pronounced HAART-induced rise of CD4 T-cell count
infecting parasite species and the immune response of the should be achieved in order to gain control of the infection.
host [45]. In HIV-infected patients, the presentation of VL Several studies using quantitative or semi-quantitative
is not significantly different from that observed in HIV- polymerase chain reaction (PCR) monitoring of parasitemia
uninfected individuals. In fact, the classic features of VL in HIV-infected patients have demonstrated low-level
continuous persistence of Leishmania DNAemia, in spite
of adequate clinical response to specific therapy [47, 48].
Bourgeois et al. proposed that the above-mentioned
condition, characterized by alternating asymptomatic and
symptomatic periods, be recognized as a new clinical entity
that the authors name “active chronic visceral leishmania-
sis” [49]. In addition, post-kala-azar dermal leishmaniasis
(PKDL), a cutaneous entity observed in patients with a
previous history of treated VL and generally considered to
be confined to the Indian subcontinent and Sudan, has been
recognized as a manifestation of anti-Leishmania immune
reconstitution inflammatory syndrome (IRIS) in HIV-
positive patients following the administration of antiretro-
viral therapy [50]. A single case of diffuse cutaneous
leishmaniasis caused by L. chagasi was also described in a
Nicaraguan patient with AIDS as IRIS [51].
Fig. 1 Cutaneous leishmaniasis “wet form” caused by Leishmania Among solid organ transplant recipients, fever and
major in an Italian soldier deployed in Afghanistan pancytopenia are the main clinical manifestations of VL
Eur J Clin Microbiol Infect Dis (2012) 31:109–118 113

[31]; since the clinical picture may simulate other infec- tion of parasite DNA with an analytical sensitivity of
tions, for those patients living outside endemic areas, a around 1 fg, which corresponds to 0.01 parasites per
careful travel history should be elicited. In general, the reaction [60]. Leishmania parasites are rare and hard to
onset of VL has been observed as a late complication of visualize in skin lesions of patients affected by PKDL with
transplantation, but in a recent series of kidney recipients, it macular/papular presentation, making it extremely difficult
was diagnosed in the early (median 17 days) post-transplant to differentiate PKDL from leprosy by traditional methods.
period [32]. Kidney involvement by the parasite with the However, real-time PCR readily detected Leishmania DNA
development of nephritis or the occlusion of renal vascu- in all 25 cutaneous samples of patients with PKDL [60].
lature and subsequent graft dysfunction has also been Using quantitation by the real-time PCR of 7SL RNA gene
recently reported [52, 53]. As observed among HIV- expression levels, Romero et al. provided proof that viable
infected patients, multiple episodes of VL relapse can occur parasites are present in extralesional sites (including normal
in kidney transplant recipients, sometimes with atypical skin, tonsils, and blood monocytes) of patients with dermal
mucosal or cutaneous presentations [53]. leishmaniasis [61].
Insightful reviews on CL have recently appeared in the Direct agglutination (DAT) and immunochromato-
literature, to which the readers are referred for additional graphic (ICT) strip tests using the k39 antigen have become
information [41, 54, 55]. Interestingly, Akilov et al. popular in Africa and the Indian subcontinent for the rapid
reviewed the known morphologic types of CL and and sensitive diagnosis of Leishmania infection in the field,
proposed a new classification based on pathogenesis and with the latter being more user-friendly [62]. However, the
histologic picture [55]. These authors distinguish between k39Ag test was found to be less reliable when used in East
exogenous (localized and disseminated) and endogenous Africa than in the Indian subcontinent, with sensitivities
CL (recurrent or lupoid and PKDL). Localized cutaneous and specificities of, respectively, 75% versus 85% and 70%
leishmaniasis (LCL) is divided into typical (with lesions versus 92% [63]. A newly developed assay based on the
appearing as papule, nodule, plaque, nodulo-ulcerative, and detection of the k28 fusion protein has shown, in studies
impetiginous) and atypical forms. The latter may vary in performed in Sudan and Bangladesh, a sensitivity of,
morphology (i.e., zosteriform, eczematoid, erysipeloid, respectively, 96% and 98% [64]. If these preliminary results
verrucoid, psoriasiform) or localization (fingers, palms can be confirmed in large-scale field evaluations, this novel
and soles, lips, eyelids, and genitals). Disseminated assay could be implemented as a first-line, point-of-care test
cutaneous leishmaniasis (DCL) is distinguished from for the diagnosis of Leishmania infection.
diffuse (or pseudolepromatous) CL by the presence, in the
former, of subcutaneous nodules. The presentation with
multiple lesions may be the consequence of multiple Treatment
inoculations or secondary to the dissemination of the
parasite mediated by circulating immune cells or represent Liposomal amphotericin B (L-AMB) is, at present, unambig-
an allergic reaction (sometimes triggered by therapy), with uously considered to be the drug of choice for the treatment of
the appearance of leishmanids. Although parasite dissem- VL in high-income countries, but its prohibitively high cost
ination during CL is generally observed in patients affected precludes its widespread use in low-income, highly endemic
by L. major and L. tropica infections, a multifocal form of countries. However, the preferential price offered to the
CL caused by L. infantum has been recently described in governments of these areas, together with the recent imple-
the Mediterranean basin [56]. Finally, in South American mentation of short-course, high-dose regimens, make this
HIV-positive patients, a high prevalence of tegumentary option attractive also for developing countries [65–67].
leishmaniasis with atypical presentations (53% mucocuta- Recently, in a non-inferiority study conducted in Bihar, it
neous) or unusual localizations (27% in the genital areas) was shown that a single, high dose of L-AMB (i.e., 10 mg/
has been described [57] . kg of body weight) is as effective as, but less expensive than,
15 alternate-day infusions of amphotericin B deoxycholate at
the dose of 1 mg/kg of body weight (cure rates at 1 and
Diagnosis 6 months of 100% and 95.7% for the former and 98% and
96.3% for the latter, respectively) [65]. This study extends a
Although a standardized, commercially available PCR previous experience by the same group in which different
assay for Leishmania diagnosis is presently still unavail- dosages of L-AMB were used and another investigation
able, it is well accepted that PCR can be applied to different conducted in Greece, where two single infusions of L-AMB
clinical samples with higher sensitivities than conventional at a dosage of 10 mg/kg of body weight were employed [66–
parasitological methods for the diagnosis of VL and CL 68]. Moreover, it is worth noting that this high dosage of L-
[58, 59]. Real-time PCR allows the consistent quantifica- AMB was not associated with an increase of adverse events.
114 Eur J Clin Microbiol Infect Dis (2012) 31:109–118

Miltefosine, a phosphatidylcholine analog, is the only Bolivia showed that the 6-month response rate induced by
oral treatment licensed in India and Germany for the miltefosine in patients infected by L.(V.) braziliensis was
therapy of VL and in Colombia and Bolivia for CL. In a statistically similar to that obtained with intramuscular
phase IV open-label trial conducted in India in more than antimony (88% vs. 94%), although antimony achieved
1,100 adult and pediatric patients with VL, miltefosine clinical cure more rapidly [74]. More recently, an open-label
showed an efficacy rate of 82% in the intention-to-treat randomized study conducted in Colombia demonstrated a
analysis and of 95% in the per protocol analysis [69]. The higher response rate (both in the per protocol and intention-to-
drug was well tolerated, with diarrhea and vomiting being treat analyses) in patients treated with meglumine antimoniate
the most common adverse events observed in 8% of versus those treated with miltefosine, without any association
patients during the first week of treatment. A randomized emerging between drug efficacy and infecting Leishmania
controlled trial conducted in both HIV-positive and HIV- species [75]. Experiences on the treatment with miltefosine
negative adult Ethiopian males compared miltefosine of Old World CL caused by L. major and L. tropica are quite
(100 mg/kg day) with standard antimony therapy: the cure limited, but they seem to support an activity of the drug also
rate at 6 months were similar in HIV-negative patients (77% for these infections [73, 76, 79].
vs. 76%) but were lower in HIV-positive subjects (46% vs. The best treatment approach for CL is, at present, still a
57%), although statistical significance was not achieved matter of debate. A recent systematic analysis of published
[70]. Miltefosine was previously used in a compassionate studies included in two Cochrane reviews evidenced
program in 39 HIV-coinfected patients with VL: 41% several flaws in the criteria of randomization, the identifi-
achieved initial cure and 23% improved upon a second cation of causative species, adopted cure criteria, and time
course of treatment [71]. Several trials have employed of follow-up, making it difficult to draw any conclusive
miltefosine in comparison with placebo or antimonials for indication [80].
the treatment of Old and New World CL (Table 2) [72–78]. A systematic review of 22 articles on the treatment of
In a double-blind, placebo-controlled trial, a marked mucosal leishmaniasis in Latin America showed statistical-
difference in response rates was initially noted according ly significant better cure rates with meglumine antimoniate
to the country and Leishmania species involved. Unsatis- in comparison with stibogluconate (88% vs. 51%, p<0.05),
factory responses were evidenced in patients infected by whereas pentamidine and amphotericin B deoxycholate
L.(V.) braziliensis in Guatemala, while the contrary was were found to be as effective as meglumine [81].
recorded in individuals infected by L.(V.) panamensis in Paromomycin has been registered for VL treatment in
Colombia [72]. However, a subsequent study conducted in India after a multicenter phase III study demonstrated that

Table 2 Experience on the use of oral miltefosine for the treatment of cutaneous leishmaniasis (CL)

Reference, year Type of study Drug(s) and dosage/duration Leishmania spp. (country) Response rate at Response rate at
6 months PP 6 months ITT

[72], 2004 Double-blind, Miltefosine 2.5 mg/kg/d vs. L.(V.) panamensis (Colombia) 40/44 (91%) 82%
placebo-controlled placebo/28 days L.(V.) braziliensis (Guatemala) 20/38 (53%) 50%
[73], 2007 Randomized, open-label Miltefosine 150 mg d for 28 days L. major (Iran) 26/28 (92.9%) 81.3%
comparative (1:1) vs. meglumine antimoniate 25/31 (83.3%) 80.6%
20 mg/kg/d for 14 days
[74], 2008 Randomized, open-label Miltefosine 2.5 mg/kg/d for L.(V.) braziliensis (Bolivia) 36/41 (88%) NR
comparative (2:1) 28 days vs. meglumine 15/16 (94%) NR
antimoniate 20 mg/kg/d for
20 days
[75], 2010 Randomized, open-label Miltefosine 150 mg d for 28 days L.(V.) braziliensis 85/122 (69.8%) 85/145 (58.6%)
comparative (1:1) vs. meglumine antimoniate (Colombia)
20 mg/kg/d for 20 days L.(V.) panamensis 103/121 (85.1%) 103/143 (72%)
[76], 2010 Observational Miltefosine 150 mg d for 28 days L. major (Afghanistan) 30/34 (88.2%) NR
retrospective
[77], 2010 Randomized, open-label Miltefosine 2.5 mg/kg/d vs. L.(V.) braziliensis (Brazil) 45/60 (75%) NR
meglumine antimoniate 18/30 (53.3%)
20 mg/kg/d for 20 days
[78], 2011 Randomized, open-label Miltefosine 2.5 mg/kg/d vs. L. (V.) guyanensis (Brazil) 71.4% NR
comparative (2:1) meglumine antimoniate 53.6%
20 mg/kg/d for 20 days

PP = per protocol; ITT = intention-to-treat; NR= not reported


Eur J Clin Microbiol Infect Dis (2012) 31:109–118 115

this drug, at the dosage of 15 mg/kg/d for 21 days, is safe, Conclusions


affordable, and efficacious [82]. However, in a multicenter
phase III trial conducted in East Africa (Sudan, Kenya, and The last 10 years have witnessed an extraordinary progress
Ethiopia) comparing three treatment regimens (paromomy- in our capacity in classifying, diagnosing, and treating
cin, sodium stibogluconate, and a combination of both), the Leishmania infection. The challenge is to translate such
paromomycin arm had to be discontinued due to poor progress in effective strategy toward programs aimed to
efficacy, especially in Sudan [83]. Although sodium control and eliminate the disease.
stibogluconate remains an effective treatment in this area,
there is growing evidence that it is associated with a higher
mortality risk among patients older than 45 years [84]. References
Given that, in East Africa, miltefosine is neither registered
nor available and L-AMB remains an unaffordable drug, 1. Al-Mutawakel K, Scutaru C, Shami A, Sakr M, Groneberg
there was a rationale to pursue studies employing paromo- DA, Quarcoo D (2010) Scientometric analysis of the world-
mycin. A phase II dose-finding study conducted in Sudan wide research efforts concerning Leishmaniasis. Parasit Vectors
3:14
showed that, in fact, increasing the dose of paromomycin
2. Pratt DM, David JR (1981) Monoclonal antibodies that distin-
(20 mg/kg/d for 21 days) or extending treatment duration guish between New World species of Leishmania. Nature
(15 mg/kg/d for 28 days) improves efficacy of this drug to 291:581–583
80–81% cure rates at 6 months, a figure that is closer to 3. Lainson R, Shaw JJ (1987) Evolution, classification and geo-
graphical distribution. In: Peters W, Killick-Kendrick R (eds) The
what was originally observed in India [85]. Parenteral and leishmaniases in biology and medicine. Academic Press, London,
topical paromomycin has also been used for the treatment pp 1–120
of CL, but with cure rates ranging from 4 to 93%. A recent 4. Rioux JA, Lanotte G, Serres E, Pratlong F, Bastien P, Perieres J
meta-analysis of 14 randomized controlled trials that (1990) Taxonomy of Leishmania. Use of isoenzymes. Suggestions
for a new classification. Ann Parasitol Hum Comp 65:111–125
included 1,221 patients showed that topical paromomycin
5. Bañuls AL, Hide M, Prugnolle F (2007) Leishmania and the
administration has therapeutic activity against Old and New leishmaniases: a parasite genetic update and advances in taxon-
World CL [86]. Moreover, when compared with pentava- omy, epidemiology and pathogenicity in humans. Adv Parasitol
lent antimony compounds, topical paromomycin had a 64:1–109
6. Lukes J, Mauricio IL, Schönian G, Dujardin JC, Soteriadou K,
similar efficacy to intralesionally administered antimonials
Dedet JP, Kuhls K, Tintaya KW, Jirků M, Chocholová E,
in Old World CL, but was inferior to parenteral antimonials Haralambous C, Pratlong F, Oborník M, Horák A, Ayala FJ,
in treating New World CL [86]. However, no significant Miles MA (2007) Evolutionary and geographical history of the
differences in efficacy emerged when parenteral paromo- Leishmania donovani complex with a revision of current
taxonomy. Proc Natl Acad Sci USA 104:9375–9380
mycin was compared to parenteral antimonials in the
7. Fraga J, Montalvo AM, De Doncker S, Dujardin JC, Van der
treatment of New World CL. Auwera G (2010) Phylogeny of Leishmania species based on the
Finally, recent years have witnessed a growing interest heat-shock protein 70 gene. Infect Genet Evol 10:238–245
in exploring the use of combination therapy also for the 8. Schönian G, Mauricio I, Cupolillo E (2010) Is it time to revise the
nomenclature of Leishmania? Trends Parasitol 26:466–469
treatment of VL in order to: achieve shorter treatment
9. Bern C, Maguire JH, Alvar J (2008) Complexities of assessing the
duration; reduce the total drug dose administered; limit disease burden attributable to leishmaniasis. PLoS Negl Trop Dis
the emergence of drug resistance; and enhance the 2:e313
clinical and parasitological cure rates in difficult-to-treat 10. Singh SP, Reddy DC, Rai M, Sundar S (2006) Serious under-
reporting of visceral leishmaniasis through passive case reporting
populations, such as HIV-infected patients [87, 88]. An
in Bihar, India. Trop Med Int Health 11:899–905
analysis of the cost-effectiveness of Leishmania combina- 11. Singh VP, Ranjan A, Topno RK, Verma RB, Siddique NA,
tion therapy for the Indian subcontinent shows that the Ravidas VN, Kumar N, Pandey K, Das P (2010) Estimation of
association of miltefosine with paromomycin represents under-reporting of visceral leishmaniasis cases in Bihar, India. Am
J Trop Med Hyg 82:9–11
the most economically attractive strategy, with a cost of
12. Das P, Samuels S, Desjeux P, Mittal A, Topno R, Siddiqui NA,
US$92 per single averted death, followed by liposomal Sur D, Pandey A, Sarnoff R (2010) Annual incidence of visceral
amphotericin B plus paromomycin (US$652 per single leishmaniasis in an endemic area of Bihar, India. Trop Med Int
averted death) [89]. A phase III non-inferiority, open-label Health 15(Suppl 2):4–11
13. Dujardin JC, Campino L, Cañavate C, Dedet JP, Gradoni L,
trial conducted in India comparing standard treatment
Soteriadou K, Mazeris A, Ozbel Y, Boelaert M (2008) Spread of
(amphotericin B infusion on alternate days for 30 days) vector-borne diseases and neglect of Leishmaniasis, Europe.
with three drug combinations (single-dose L-AMB and Emerg Infect Dis 14:1013–1018
10-day paromomycin; 10 days each of miltefosine and 14. Ready PD (2010) Leishmaniasis emergence in Europe. Euro
Surveill 15:19505
paromomycin; single dose L-AMB and 7-day miltefosine)
15. Maroli M, Rossi L, Baldelli R, Capelli G, Ferroglio E, Genchi C,
showed cure rates at 6 months of at least 97% for all Gramiccia M, Mortarino M, Pietrobelli M, Gradoni L (2008) The
combinations [90]. northward spread of leishmaniasis in Italy: evidence from
116 Eur J Clin Microbiol Infect Dis (2012) 31:109–118

retrospective and ongoing studies on the canine reservoir and 33. Weisser M, Khanlari B, Terracciano L, Arber C, Gratwohl A,
phlebotomine vectors. Trop Med Int Health 13:256–264 Bassetti S, Hatz C, Battegay M, Flückiger U (2007) Visceral
16. Morosetti G, Bongiorno G, Beran B, Scalone A, Moser J, leishmaniasis: a threat to immunocompromised patients in non-
Gramiccia M, Gradoni L, Maroli M (2009) Risk assessment for endemic areas? Clin Microbiol Infect 13:751–753
canine leishmaniasis spreading in the north of Italy. Geospat 34. Xynos ID, Tektonidou MG, Pikazis D, Sipsas NV (2009)
Health 4:115–127 Leishmaniasis, autoimmune rheumatic disease, and anti-tumor
17. Biglino A, Bolla C, Concialdi E, Trisciuoglio A, Romano A, necrosis factor therapy, Europe. Emerg Infect Dis 15:956–959
Ferroglio E (2010) Asymptomatic Leishmania infantum infection 35. Cascio A, Iaria M, Iaria C (2010) Leishmaniasis and biologic
in an area of Northwestern Italy (Piedmont Region) where such therapies for rheumatologic diseases. Semin Arthritis Rheum 40:
infections are traditionally nonendemic. J Clin Microbiol 48:131– e3–e5
136 36. De Leonardis F, Govoni M, Lo Monaco A, Trotta F (2009)
18. Naucke TJ, Schmitt C (2004) Is leishmaniasis becoming endemic Visceral leishmaniasis and anti-TNF-α therapy: case report and
in Germany? Int J Med Microbiol 293:179–181 review of the literature. Clin Exp Rheumatol 27:503–506
19. Naucke TJ, Menn B, Massberg D, Lorentz S (2008) Sandflies and 37. Mueller MC, Fleischmann E, Grunke M, Schewe S, Bogner JR,
leishmaniasis in Germany. Parasitol Res 103(Suppl 1):S65–S68 Löscher T (2009) Relapsing cutaneous leishmaniasis in a patient
20. Postigo JA (2010) Leishmaniasis in the World Health Organiza- with ankylosing spondylitis treated with infliximab. Am J Trop
tion Eastern Mediterranean Region. Int J Antimicrob Agents 36 Med Hyg 81:52–54
(Suppl 1):S62–S65 38. Kopterides P, Mourtzoukou EG, Skopelitis E, Tsavaris N, Falagas
21. Mosleh IM, Geith E, Natsheh L, Abdul-Dayem M, Abotteen N ME (2007) Aspects of the association between leishmaniasis and
(2008) Cutaneous leishmaniasis in the Jordanian side of the malignant disorders. Trans R Soc Trop Med Hyg 101:1181–1189
Jordan Valley: severe under-reporting and consequences on public 39. Antinori S, Gianelli E, Calattini S, Longhi E, Gramiccia M,
health management. Trop Med Int Health 13:855–860 Corbellino M (2005) Cutaneous leishmaniasis: an increasing
22. Romero GA, Boelaert M (2010) Control of visceral leishmaniasis threat for travellers. Clin Microbiol Infect 11:343–346
in Latin America—a systematic review. PLoS Negl Trop Dis 4: 40. Pavli A, Maltezou HC (2010) Leishmaniasis, an emerging
e584 infection in travellers. Int J Infect Dis 14:e1032–e1039
23. Alvar J, Cañavate C, Gutiérrez-Solar B, Jiménez M, Laguna F, 41. Schwartz E, Hatz C, Blum J (2006) New world cutaneous
López-Vélez R, Molina R, Moreno J (1997) Leishmania and leishmaniasis in travellers. Lancet Infect Dis 6:342–349
human immunodeficiency virus coinfection: the first 10 years. 42. Weina PJ, Neafie RC, Wortmann G, Polhemus M, Aronson NE
Clin Microbiol Rev 10:298–319 (2004) Old world leishmaniasis: an emerging infection among
24. Kubar J, Marty P, Lelièvre A, Quaranta JF, Staccini P, Caroli- deployed US military and civilian workers. Clin Infect Dis
Bosc C, Le Fichoux Y (1998) Visceral leishmaniosis in HIV- 39:1674–1680
positive patients: primary infection, reactivation and latent 43. van Thiel PP, Leenstra T, de Vries HJ, van der Sluis A, van Gool
infection: impact of the CD4+ T-lymphocyte counts. AIDS T, Krull AC, van Vugt M, de Vries PJ, Zeegelaar JE, Bart A, van
12:2147–2153 der Meide WF, Schallig HD, Faber WR, Kager PA (2010)
25. Paredes R, Munoz J, Diaz I, Domingo P, Gurgui M, Clotet B Cutaneous leishmaniasis (Leishmania major infection) in Dutch
(2003) Leishmaniasis in HIV infection. J Postgrad Med 49:39–49 troops deployed in northern Afghanistan: epidemiology, clinical
26. Alvar J, Aparicio P, Aseffa A, Den Boer M, Cañavate C, Dedet JP, aspects, and treatment. Am J Trop Med Hyg 83:1295–1300
Gradoni L, Ter Horst R, López-Vélez R, Moreno J (2008) The 44. Lederman ER, Weld LH, Elyazar IRF, von Sonnenburg F, Loutan
relationship between leishmaniasis and AIDS: the second 10 years. L, Schwartz E, Keystone JS (2008) Dermatologic conditions of
Clin Microbiol Rev 21:334–359 the ill returned traveler: an analysis from the GeoSentinel
27. de la Rosa R, Pineda JA, Delgado J, Macías J, Morillas F, Mira Surveillance Network. Int J Infect Dis 12:593–602
JA, Sánchez-Quijano A, Leal M, Lissen E (2002) Incidence of and 45. Pintado V, Martín-Rabadán P, Rivera ML, Moreno S, Bouza E
risk factors for symptomatic visceral leishmaniasis among human (2001) Visceral leishmaniasis in human immunodeficiency virus
immunodeficiency virus type 1-infected patients from Spain in the (HIV)-infected and non-HIV-infected patients. A comparative
era of highly active antiretroviral therapy. J Clin Microbiol study. Medicine (Baltimore) 80:54–73
40:762–767 46. López-Vélez R (2003) The impact of highly active antiretroviral
28. ter Horst R, Collin SM, Ritmeijer K, Bogale A, Davidson RN therapy (HAART) on visceral leishmaniasis in Spanish patients
(2008) Concordant HIV infection and visceral leishmaniasis in who are co-infected with HIV. Ann Trop Med Parasitol 97(Suppl
Ethiopia: the influence of antiretroviral treatment and other factors 1):143–147
on outcome. Clin Infect Dis 46:1702–1709 47. Mary C, Faraut F, Lascombe L, Dumon H (2004) Quantification
29. Gorski S, Collin SM, Ritmeijer K, Keus K, Gatluak F, Mueller M, of Leishmania infantum DNA by a real-time PCR assay with high
Davidson RN (2010) Visceral leishmaniasis relapse in Southern sensitivity. J Clin Microbiol 42:5249–5255
Sudan (1999–2007): a retrospective study of risk factors and 48. Antinori S, Calattini S, Piolini R, Longhi E, Bestetti G, Cascio A,
trends. PLoS Negl Trop Dis 4:e705 Parravicini C, Corbellino M (2009) Is real-time polymerase chain
30. Petrela R, Kuneshka L, Foto E, Zavalani F, Gradoni L (2010) reaction (PCR) more useful than a conventional PCR for the
Pediatric visceral leishmaniasis in Albania: a retrospective clinical management of leishmaniasis? Am J Trop Med Hyg
analysis of 1,210 consecutive hospitalized patients (1995–2009). 81:46–51
PLoS Negl Trop Dis 4:e814 49. Bourgeois N, Bastien P, Reynes J, Makinson A, Rouanet I,
31. Antinori S, Cascio A, Parravicini C, Bianchi R, Corbellino M Lachaud L (2010) ‘Active chronic visceral leishmaniasis’ in HIV-
(2008) Leishmaniasis among organ transplant recipients. Lancet 1-infected patients demonstrated by biological and clinical long-
Infect Dis 8:191–199 term follow-up of 10 patients. HIV Med 11:670–673
32. Veroux M, Corona D, Giuffrida G, Cacopardo B, Sinagra N, 50. Antinori S, Longhi E, Bestetti G, Piolini R, Acquaviva V, Foschi
Tallarita T, Giaquinta A, Zerbo D, Veroux PF (2010) Visceral A, Trovati S, Parravicini C, Corbellino M, Meroni L (2007) Post-
leishmaniasis in the early post-transplant period after kidney kala-azar dermal leishmaniasis as an immune reconstitution
transplantation: clinical features and therapeutic management. inflammatory syndrome in a patient with acquired immune
Transpl Infect Dis 12:387–391 deficiency syndrome. Br J Dermatol 157:1032–1036
Eur J Clin Microbiol Infect Dis (2012) 31:109–118 117

51. Sinha S, Fernández G, Kapila R, Lambert WC, Schwartz RA 67. Sundar S, Jha TK, Thakur CP, Mishra M, Singh VP, Buffels R
(2008) Diffuse cutaneous leishmaniasis associated with the (2003) Single-dose liposomal amphotericin B in the treatment of
immune reconstitution inflammatory syndrome. Int J Dermatol visceral leishmaniasis in India: a multicenter study. Clin Infect Dis
47:1263–1270 37:800–804
52. Dettwiler S, McKee T, Hadaya K, Chappuis F, van Delden C, 68. Syriopoulou V, Daikos GL, Theodoridou M, Pavlopoulou I,
Moll S (2010) Visceral leishmaniasis in a kidney transplant Manolaki AG, Sereti E, Karamboula A, Papathanasiou D, Krikos
recipient: parasitic interstitial nephritis, a cause of renal dysfunc- X, Saroglou G (2003) Two doses of a lipid formulation of
tion. Am J Transpl 10:1486–1489 amphotericin B for the treatment of Mediterranean visceral
53. Simon I, Wissing KM, Del Marmol V, Antinori S, Remmelink M, leishmaniasis. Clin Infect Dis 36:560–566
Nilufer Broeders E, Nortier JL, Corbellino M, Abramowicz D, 69. Bhattacharya SK, Sinha PK, Sundar S, Thakur CP, Jha TK,
Cascio A (2011) Recurrent leishmaniasis in kidney transplant Pandey K, Das VR, Kumar N, Lal C, Verma N, Singh VP, Ranjan
recipients: report of 2 cases and systematic review of the literature. A, Verma RB, Anders G, Sindermann H, Ganguly NK (2007)
Transpl Infect Dis. doi:10.1111/j.1399-3062.2011.00598.x Phase 4 trial of miltefosine for the treatment of Indian visceral
54. Reithinger R, Dujardin JC, Louzir H, Pirmez C, Alexander B, leishmaniasis. J Infect Dis 196:591–598
Brooker S (2007) Cutaneous leishmaniasis. Lancet Infect Dis 70. Ritmeijer K, Dejenie A, Assefa Y, Hundie TB, Mesure J, Boots G,
7:581–596 den Boer M, Davidson RN (2006) A comparison of miltefosine
55. Akilov OE, Khachemoune A, Hasan T (2007) Clinical manifes- and sodium stibogluconate for treatment of visceral leishmaniasis
tations and classification of Old World cutaneous leishmaniasis. in an Ethiopian population with high prevalence of HIV infection.
Int J Dermatol 46:132–142 Clin Infect Dis 43:357–364
56. Maniscalco M, Noto G, Zichichi L, Veraldi S (2007) Multifocal 71. Sindermann H, Engel KR, Fischer C, Bommer W (2004) Oral
cutaneous leishmaniasis: a new clinical presentation of the miltefosine for leishmaniasis in immunocompromised patients:
disease. Acta Derm Venereol 87:275–276 compassionate use in 39 patients with HIV infection. Clin Infect
57. Lindoso JA, Barbosa RN, Posada-Vergara MP, Duarte MI, Dis 39:1520–1523
Oyafuso LK, Amato VS, Goto H (2009) Unusual manifestations 72. Soto J, Arana BA, Toledo J, Rizzo N, Vega JC, Diaz A, Luz M,
of tegumentary leishmaniasis in AIDS patients from the New Gutierrez P, Arboleda M, Berman JD, Junge K, Engel J,
World. Br J Dermatol 160:311–318 Sindermann H (2004) Miltefosine for New World cutaneous
58. Antinori S, Calattini S, Longhi E, Bestetti G, Piolini R, Magni C, leishmaniasis. Clin Infect Dis 38:1266–1272
Orlando G, Gramiccia M, Acquaviva V, Foschi A, Corvasce S, 73. Mohebali M, Fotouhi A, Hooshmand B, Zarei Z, Akhoundi B,
Colomba C, Titone L, Parravicini C, Cascio A, Corbellino M Rahnema A, Razaghian AR, Kabir MJ, Nadim A (2007)
(2007) Clinical use of polymerase chain reaction performed on Comparison of miltefosine and meglumine antimoniate for the
peripheral blood and bone marrow samples for the diagnosis and treatment of zoonotic cutaneous leishmaniasis (ZCL) by a
monitoring of visceral leishmaniasis in HIV-infected and HIV- randomized clinical trial in Iran. Acta Trop 103:33–40
uninfected patients: a single-center, 8-year experience in Italy and 74. Soto J, Rea J, Balderrama M, Toledo J, Soto P, Valda L, Berman
review of the literature. Clin Infect Dis 44:1602–1610 JD (2008) Efficacy of miltefosine for Bolivian cutaneous
59. Bensoussan E, Nasereddin A, Jonas F, Schnur LF, Jaffe CL (2006) leishmaniasis. Am J Trop Med Hyg 78:210–211
Comparison of PCR assays for diagnosis of cutaneous leishman- 75. Vélez I, López L, Sánchez X, Mestra L, Rojas C, Rodríguez E
iasis. J Clin Microbiol 44:1435–1439 (2010) Efficacy of miltefosine for the treatment of American
60. Verma S, Kumar R, Katara GK, Singh LC, Negi NS, Ramesh V, cutaneous leishmaniasis. Am J Trop Med Hyg 83:351–356
Salotra P (2010) Quantification of parasite load in clinical samples 76. van Thiel PP, Leenstra T, Kager PA, de Vries HJ, van Vugt M, van
of leishmaniasis patients: IL-10 level correlates with parasite load der Meide WF, Bart A, Zeegelaar JE, van der Sluis A, Schallig
in visceral leishmaniasis. PLoS One 5:e10107 HD, van Gool T, Faber WR, de Vries PJ (2010) Miltefosine
61. Romero I, Téllez J, Suárez Y, Cardona M, Figueroa R, Zelazny A, treatment of Leishmania major infection: an observational study
Gore Saravia N (2010) Viability and burden of Leishmania in involving Dutch military personnel returning from northern
extralesional sites during human dermal leishmaniasis. PLoS Negl Afghanistan. Clin Infect Dis 50:80–83
Trop Dis 4:e819 77. Machado PR, Ampuero J, Guimarães LH, Villasboas L, Rocha
62. Srivastava P, Dayama A, Mehrotra S, Sundar S (2011) Diagnosis AT, Schriefer A, Sousa RS, Talhari A, Penna G, Carvalho EM
of visceral leishmaniasis. Trans R Soc Trop Med Hyg 105:1–6 (2010) Miltefosine in the treatment of cutaneous leishmaniasis
63. Boelaert M, El-Safi S, Hailu A, Mukhtar M, Rijal S, Sundar S, caused by Leishmania braziliensis in Brazil: a randomized and
Wasunna M, Aseffa A, Mbui J, Menten J, Desjeux P, Peeling controlled trial. PLoS Negl Trop Dis 4:e912
RW (2008) Diagnostic tests for Kala-azar: a multi-centre study 78. Chrusciak-Talhari A, Dietze R, Chrusciak Talhari C, da Silva RM,
of the freeze-dried DAT, rK39 strip test and KAtex in East Gadelha Yamashita EP, de Oliveira Penna G, Lima Machado PR,
Africa and the Indian subcontinent. Trans R Soc Trop Med Talhari S (2011) Randomized controlled clinical trial to assess
Hyg 102:32–40 efficacy and safety of miltefosine in the treatment of cutaneous
64. Pattabhi S, Whittle J, Mohamath R, El-Safi S, Moulton GG, leishmaniasis caused by Leishmania (Viannia) guyanensis in
Guderian JA, Colombara D, Abdoon AO, Mukhtar MM, Mondal Manaus, Brazil. Am J Trop Med Hyg 84:255–260
D, Esfandiari J, Kumar S, Chun P, Reed SG, Bhatia A (2010) 79. Keynan Y, Larios OE, Wiseman MC, Plourde M, Ouellette M,
Design, development and evaluation of rK28-based point-of-care Rubinstein E (2008) Use of oral miltefosine for cutaneous
tests for improving rapid diagnosis of visceral leishmaniasis. leishmaniasis in Canadian soldiers returning from Afghanistan.
PLoS Negl Trop Dis 4:e822 Can J Infect Dis Med Microbiol 19:394–396
65. Sundar S, Chakravarty J, Agarwal D, Rai M, Murray HW (2010) 80. González U, Pinart M, Reveiz L, Rengifo-Pardo M, Tweed J,
Single-dose liposomal amphotericin B for visceral leishmaniasis Macaya A, Alvar J (2010) Designing and reporting clinical trials
in India. N Engl J Med 362:504–512 on treatments for cutaneous leishmaniasis. Clin Infect Dis 51:409–
66. Sundar S, Agrawal G, Rai M, Makharia MK, Murray HW (2001) 419
Treatment of Indian visceral leishmaniasis with single or daily 81. Amato VS, Tuon FF, Siqueira AM, Nicodemo AC, Neto VA
infusions of low dose liposomal amphotericin B: randomised trial. (2007) Treatment of mucosal leishmaniasis in Latin America:
BMJ 323:419–422 systematic review. Am J Trop Med Hyg 77:266–274
118 Eur J Clin Microbiol Infect Dis (2012) 31:109–118

82. Sundar S, Jha TK, Thakur CP, Sinha PK, Bhattacharya SK (2007) treatment of visceral leishmaniasis in Sudan: a randomized, open-
Injectable paromomycin for visceral leishmaniasis in India. N label, dose-finding study. PLoS Negl Trop Dis 4:e855
Engl J Med 356:2571–2581 86. Kim DH, Chung HJ, Bleys J, Ghohestani RF (2009) Is
83. Hailu A, Musa A, Wasunna M, Balasegaram M, Yifru S, paromomycin an effective and safe treatment against cutaneous
Mengistu G, Hurissa Z, Hailu W, Weldegebreal T, Tesfaye S, leishmaniasis? A meta-analysis of 14 randomized controlled trials.
Makonnen E, Khalil E, Ahmed O, Fadlalla A, El-Hassan A, PLoS Negl Trop Dis 3:e381
Raheem M, Mueller M, Koummuki Y, Rashid J, Mbui J, Mucee 87. Olliaro PL (2010) Drug combinations for visceral leishmaniasis.
G, Njoroge S, Manduku V, Musibi A, Mutuma G, Kirui F, Curr Opin Infect Dis 29:595–602
Lodenyo H, Mutea D, Kirigi G, Edwards T, Smith P, Muthami L, 88. van Griensven J, Balasegaram M, Meheus F, Alvar J, Lynen L,
Royce C, Ellis S, Alobo M, Omollo R, Kesusu J, Owiti R, Boelaert M (2010) Combination therapy for visceral leishmania-
Kinuthia J; Leishmaniasis East Africa Platform (LEAP) group sis. Lancet Infect Dis 10:184–194
(2010) Geographical variation in the response of visceral 89. Meheus F, Balasegaram M, Olliaro P, Sundar S, Rijal S, Faiz MA,
leishmaniasis to paromomycin in East Africa: a multicentre, Boelaert M (2010) Cost-effectiveness analysis of combination
open-label, randomized trial. PLoS Negl Trop Dis 4:e709 therapies for visceral leishmaniasis in the Indian subcontinent.
84. Chappuis F, Alirol E, Worku DT, Mueller Y, Ritmeijer K (2011) PLoS Negl Trop Dis 4:e818
High mortality among older patients treated with pentavalent 90. Sundar S, Sinha PK, Rai M, Verma DK, Nawin K, Alam S,
antimonials for visceral leishmaniasis in East Africa and rationale Chakravarty J, Vaillant M, Verma N, Pandey K, Kumari P, Lal CS,
for switch to liposomal amphotericin B. Antimicrob Agents Arora R, Sharma B, Ellis S, Strub-Wourgaft N, Balasegaram M,
Chemother 55:455–456 Olliaro P, Das P, Modabber F (2011) Comparison of short-course
85. Musa AM, Younis B, Fadlalla A, Royce C, Balasegaram M, multidrug treatment with standard therapy for visceral leishmaniasis
Wasunna M, Hailu A, Edwards T, Omollo R, Mudawi M, in India: an open-label, non-inferiority, randomised controlled trial.
Kokwaro G, El-Hassan A, Khalil E (2010) Paromomycin for the Lancet 377:477–486

You might also like