Issue |
Parasite
Volume 32, 2025
|
|
---|---|---|
Article Number | 18 | |
Number of page(s) | 17 | |
DOI | https://doi.org/10.1051/parasite/2025009 | |
Published online | 05 March 2025 |
Review Article
Leishmaniases: Strategies in treatment development
Leishmanioses : stratégies de développement de traitements
1
Université Côte d’Azur, INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), BP 23194, 06204 Nice Cedex 3, France
2
Service de Parasitologie-Mycologie, Centre Hospitalier Universitaire de Nice, Hôpital de l’Archet, CS 23079, 06202 Nice Cedex 3, France
* Corresponding author: pomares.c@chu-nice.fr
Received:
5
July
2024
Accepted:
4
February
2025
Leishmaniases are vector-borne parasitic diseases that pose a threat to over 1 billion people worldwide. The parasites target cells of the reticulohistiocytic system, such as macrophages, where they replicate. The disease manifests in various forms, ranging from localized cutaneous leishmaniasis to life-threatening visceral forms, which are fatal in 95% of cases without treatment. Current treatments rely on the invasive administration of toxic and expensive drugs that are increasingly encountering resistance. Therefore, finding alternative treatments for this disease is imperative. This literature review focuses on recent advancements in alternative treatments and aims to present the various strategies designed to address current limitations, including cost, toxicity, off-target effects, administration routes, and the emergence of drug resistance. Starting with an overview of the existing approved treatments and their specific limitations, we categorize treatment development strategies into five key sections: (i) combination therapies using existing approved treatments to enhance efficacy and reduce resistance; (ii) nanoparticle formulations, which enable targeted delivery to infected organs and improved therapeutic efficiency; (iii) drug repositioning, a strategy that has already contributed to the approval of over half of current therapeutic compounds; (iv) immunomodulation, used in conjunction with standard chemotherapies to enhance treatment efficacy and lower relapse rates; and (v) ethnobotanicals, which have demonstrated promising in vitro results by combining low toxicity, immunomodulatory properties, and potent anti-parasitic effects. In summary, this review outlines current strategies in treatment development, emphasizing their advantages over conventional therapies while acknowledging their limitations.
Résumé
Les leishmanioses sont des maladies parasitaires à transmission vectorielle qui menacent plus d’un milliard de personnes dans le monde. Les parasites ciblent les cellules du système réticulohistiocytaire, comme les macrophages, où ils se répliquent. La maladie se manifeste sous diverses formes, allant de la leishmaniose cutanée localisée aux formes viscérales potentiellement mortelles, qui sont responsables de décès dans 95% des cas sans traitement. Les traitements actuels reposent sur l’administration invasive de médicaments toxiques et coûteux qui rencontrent de plus en plus de résistances. Il est donc impératif de trouver des traitements alternatifs pour cette maladie. Cette revue de la littérature se concentre sur les avancées récentes en matière de traitements alternatifs et vise à présenter les différentes stratégies conçues pour remédier aux limitations actuelles, notamment le coût, la toxicité, les effets hors cible, les voies d’administration et l’émergence de la résistance aux médicaments. En commençant par un aperçu des traitements approuvés existants et de leurs limitations spécifiques, nous classons les stratégies de développement de traitements en cinq sections clés : (i) les thérapies combinées utilisant les traitements approuvés existants pour améliorer l’efficacité et réduire la résistance; (ii) les formulations de nanoparticules, qui permettent une administration ciblée aux organes infectés et une efficacité thérapeutique améliorée; (iii) le repositionnement des médicaments, une stratégie qui a déjà contribué à l’approbation de plus de la moitié des composés thérapeutiques actuels; (iv) l’immunomodulation, utilisée en conjonction avec les chimiothérapies standard pour améliorer l’efficacité du traitement et réduire les taux de rechute; (v) les produits ethnobotaniques, qui ont démontré des résultats in vitro prometteurs en combinant une faible toxicité, des propriétés immunomodulatrices et un puissant effet antiparasitaire. En résumé, cette revue décrit les stratégies actuelles de développement de traitements, en soulignant leurs avantages par rapport aux thérapies conventionnelles tout en reconnaissant leurs limites.
Key words: Treatments / Leishmaniases / Immunomodulator / Ethnopharmacology
Edited by: Jérôme Depaquit
© A. Majoor et al., published by EDP Sciences, 2025
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Introduction
Leishmaniasis, a vector-borne disease transmitted through the bite of phlebotomine sand flies, poses a threat to more than 1 billion people in over 98 countries worldwide [111]. It is responsible for 20,000 to 30,000 deaths a year and has been considered an uncontrolled neglected tropical disease by the World Health Organization (WHO) since 2011 [111].
Leishmania parasites exhibit a preference for the reticulohistiocytic system in mammals, with macrophages serving as their primary host cells. Inside these cells, the parasite replicates until the macrophage ruptures, enabling the infection to spread. Over 20 species of Leishmania are known to cause disease, each exhibiting distinct tropisms that lead to outcomes ranging from asymptomatic carriage to diverse clinical manifestations. These manifestations range from self-healing skin ulcers to systemic infections characterized by hepatosplenomegaly, which can be fatal if left untreated.
Human leishmaniasis manifests in four main forms. The most common form, cutaneous leishmaniasis, causes ulcerative skin lesions and is typically associated with Leishmania (L.) major, L. tropica, or L. mexicana. Muco-cutaneous leishmaniasis (MCL) caused by L. braziliensis, L. panamensis, and L. amazonensis affects the mucous membranes of the nose and mouth, resulting in severe disfigurement as the disease progresses. Visceral leishmaniasis, the most severe form of the disease, primarily targets the liver and spleen, causing irregular fever and severe hepatosplenomegaly, with a mortality rate of 95% if left untreated. It is predominantly caused by L. donovani, which can also lead to post-kala-azar dermal leishmaniasis, a condition marked by widespread skin lesions that appear after an initial cure of visceral leishmaniasis [90]. Leishmania infantum is another species associated with visceral leishmaniasis.
The disease caused by L. infantum primarily affects dogs, which serve as the reservoir for this parasite. Non-infected sand flies can acquire the parasite during a blood meal from an infected dog and subsequently transmit it to other mammals, such as foxes, rodents, and humans. In dogs, clinical symptoms often begin with skin-related issues, such as alopecia, onychogryphosis, and eye problems including keratoconjunctivitis and uveitis. If untreated, these symptoms can progress to a visceral form, characterized by splenomegaly, kidney failure, weight loss, muscle wasting, and lethargy [77].
Currently, two vaccines are available for canine leishmaniasis caused by L. infantum: LetiFend® in Europe, and Leish-Tec® in Brazil [50]. However, no effective vaccine exists for humans [106]. Prevention in humans relies entirely on avoiding sand fly bites through the use of repellents, insecticide sprays, or mosquito nets. Once an infection occurs, treatment remains the only option. Pentavalent antimonials are commonly used as the first-line treatment in most countries. However, increasing drug resistance has become a major concern [36]. Nowadays, liposomal amphotericin B (L-AMB) is considered as the first-line treatment in high-income countries [80]. Despite their efficacy, existing treatments have significant limitations, including invasive administration, long treatment durations, high costs, and the emergence of resistance [75].
Given these challenges, there is an urgent need to develop novel alternative treatments. In this review, we discuss the limitations of current therapies and propose strategies to address these issues. Specifically, we focus on reducing toxicity, costs, and resistance, while exploring new administration routes and methods to shorten treatment durations.
Materials and methods
This review concentrated on studies conducted in vivo and in vitro on amastigotes, as these are the intracellular forms responsible for the disease. Screening on promastigotes was excluded because, although amastigote-based screening offers a high hit-to-success ratio, promastigote-based screening often fails during later stages of drug development [23]. This review highlights strategies for developing treatments against leishmaniasis, focusing on progress made between 2015 and 2023. Bibliographic research was conducted on PubMed using keywords such as leishmaniasis, treatment, cutaneous or mucocutaneous or visceral, mechanism, and combination. Over 600 articles matching these keywords were identified and categorized based on the drug development strategy they addressed. First, we reviewed current therapies validated for treating Leishmania, along with their mechanism of action and limitations. Combination therapies were then explored, emphasizing the simultaneous use of several approved compounds. We also examined two key strategies responsible for more than half of the major approved treatments: nanoparticle carriers, as seen with liposomal amphotericin B, and drug repositioning exemplified by miltefosine and paromomycin. Additionally, we investigated articles on immunotherapy with current treatments and studies on ethnobotanics as a strategy for novel drug discovery.
Results
Current treatment options and limitations
Currently, seven approved treatments are used against different forms of leishmaniasis worldwide (Table 1). Historically (Fig. 1), the first approved treatment, based on trivalent antimonials, was introduced in 1912. By 1922, these were replaced by safer pentavalent antimonials, leading to the development of today’s two approved molecules: sodium stibogluconate (SSG) and meglumine antimoniate. These drugs were highly effective until the 1970s and 1980s, when significant treatment failures and the emergence of resistant strains began to be observed [110]. Despite their efficacy, they are associated with side effects such as gastrointestinal issues (vomiting, anorexia) and cardiotoxicity. Pentamidine, approved in the 1940s, is another drug used to treat leishmaniasis. However, its use has been restricted by the WHO due to severe side effects, including diabetes mellitus, severe hypoglycemia, shock, and myocarditis. Ketoconazole and fluconazole, discovered in 1977 and 1978, respectively are treatment options for cutaneous leishmaniasis [110]. However, the development of resistance in Leishmania parasites has limited their effectiveness. In 1996, the approval of amphotericin B (AMB) addressed some resistance issues, but this drug is associated with side effects such as fever, chills, rigor, and nephrotoxicity. In 1999, L-AMB was developed to reduce toxicity. While liposomal encapsulation improved safety, it significantly increased the cost, making it less accessible in developing countries [12]. Until 2004, all treatments for visceral leishmaniasis required invasive administration via the intravenous or intramuscular routes. This made treatment difficult in some regions, where multiple hospital visits were required for injections. The approval of miltefosine in 2004 marked a breakthrough as the first oral drug for leishmaniasis. Despite its advantages, miltefosine is teratogenic, limiting its use in pregnant women, and causes gastrointestinal side effects [12]. Paromomycin, an aminoside antibiotic, became the most recent anti-leishmanial drug approved in 2006 [16, 86]. However, its use is limited due to side effects, including ototoxicity, renal toxicity, and hepatotoxicity.
![]() |
Figure 1 Current treatments and their development over time. |
Classical monotherapies used for Leishmania infection.
Today, these drugs (Table 1) remain the first-line treatments recommended by WHO guidelines for all forms of leishmaniasis. Despite their efficacy, they have significant limitations. Most treatments require invasive administration, have long treatment durations, and are associated with high costs and toxicity. Additionally, treatment failures are becoming more frequent, and resistant Leishmania isolates are increasingly reported for all approved regimens [75].
Combination therapies
Combination therapy is a promising strategy to address the limitations of current anti-leishmanial drugs. By combining two treatments with complementary or distinct mechanisms of action, this approach seeks to enhance therapeutic efficacy and potentially achieve synergistic effects. Targeting different biological pathways or stages of the parasite’s lifecycle also reduces the likelihood of developing drug resistance. For instance, Mutiso et al. demonstrated the effectiveness of combining diminazene and artesunate—two drugs with distinct mechanisms of action against L. donovani in a mouse model [66]. This combination was more effective than either drug alone, highlighting the potential for synergistic effects. It also reduced drug doses, lowering toxicity and treatment costs, particularly for expensive therapies like L-AMB [66].
Several drug combinations have been tested with success, significantly reducing treatment durations (Tables 2 and 3). In humans, the combination of 20 mg/kg SSG and 15 mg/kg paromomycin reduced treatment time from 30 days to 17 days, maintaining the same efficacy. In India, combining L-AMB with 50–100 mg/kg miltefosine or 11 mg/kg paromomycin shortened treatment duration from 31 to 7–10 days. It also reduced the L-AMB dose from 15 mg/kg over 30 days to a single 5 mg/kg injection, decreasing cost and toxicity [38, 88]. To select an appropriate treatment, one approach is to align with the WHO recommendations [110, 112]. For HIV-positive patients, a combination of 30 mg/kg L-AMB and 100 mg/day miltefosine over 25 days increased cure rates from 50% with L-AMB monotherapy (40 mg/kg) to 81% [105, 112].
Combination therapies for human leishmaniases.
Combination therapies for canine leishmaniases.
In dogs, reference treatment is a combination therapy of 100 mg/kg/day meglumine antimoniate and allopurinol or miltefosine and allopurinol [50]. Miltefosine promotes remission by sustaining the inflammatory environment harmful to the parasite [84]. However, Manna et al. reported fewer relapses with meglumine antimoniate and allopurinol compared to miltefosine alone [54]. Paromomycin showed safety in dogs, but did not significantly improve cure rates compared to the reference treatment [44].
Experimental treatments in hamsters have explored topical applications of paromomycin for cutaneous leishmaniasis in combination with meglumine antimoniate (Glucantime®) or miltefosine. These combinations, even at suboptimal doses, were more effective than monotherapy, suggesting that topical paromomycin may be a viable option for reducing parasite load [60]. Combining miltefosine and paromomycin also showed enhanced efficacy, with no observed cross-resistance development upon repeated exposure [42].
While combination therapies can improve efficacy, reduce toxicity, and lower costs, they are not always successful [8, 93, 109]. Risks include the potential amplification of the side effects of both treatments and the potential development of drug resistance. For example, L. donovani has shown resistance to certain drug combinations in vitro, as reported by García-Hernández et al. [31]. These findings highlight the need for careful monitoring and the strategic use of combination therapies to mitigate the risk of resistance. In addition to combination therapies, other strategies, such as improving drug delivery systems using nanoparticles, have shown promise. Encapsulation of anti-leishmanial treatments, such as amphotericin B, has demonstrated potential to enhance efficacy and reduce limitations.
Nanoparticle carriers
Nanoparticles are small assemblies of organic or inorganic matter, sized between 1 and 100 nm, that offer significant potential in drug delivery. Organic nanoparticles can be classified into two main subgroups: polymeric nanoparticles, such as dendrimers, and lipid-based nanoparticles, such as liposomes. Lipid-based nanoparticles, primarily composed of phospholipids, are highly biocompatible and assemble into bilayered structures due to their amphipathic properties. These structures have a hydrophobic layer and a hydrophilic interior, enabling the solubilization and delivery of both hydrophilic and hydrophobic drugs. Among nanoparticle types, liposomes are the most commonly FDA-approved for clinical use. Polymeric nanoparticles offer greater structural variability due to their customizable compositions, allowing for the insertion of compounds to target specific intracellular sites [59]. Nanoparticles have largely been studied to improve the efficacy of anti-cancer drugs [115]. In anti-leishmanial therapy, liposomes have already demonstrated effectiveness, as amphotericin B is clinically administered in its liposomal form. Beyond liposomal formulations, several approved anti-leishmanial treatments have been encapsulated in various nanoparticles and tested in experimental models, such as mice and hamsters, to improve drug efficacy (Tables 4 and 5). Encapsulating approved treatments in lipid-based nanoparticles, such as liposomes, enhances drug stability and protects them from degradation in harsh environments, such as gastrointestinal fluids [29]. This allows for oral administration of drugs such as AMB [76], paromomycin [2], and pentamidine [102]. Additionally, nanoparticles enable topical administration of AMB, meglumine antimoniate, miltefosine [100], and SSG [20]. Liposomal formulations provide controlled drug release, extending the drug’s half-life and reducing peak concentrations, thereby lowering toxicity, particularly on renal function. Liposomes, being lipid-based, naturally interact with macrophage-rich organs such as the liver and spleen, improving bioavailability while minimizing systemic toxicity [89]. Nanoparticles can be functionalized with targeting groups to direct drugs to specific cells or organs. For example, phosphatidylserine signals macrophages, targeting the liver and spleen [89], lactoferrin interacts with C-type lectin receptors on antigen-presenting cells [6], guar gum targets mannose-like receptors on macrophages [76] and chitosan promotes phagocytosis, favoring liver and spleen targeting [101].
Nanoparticle carriers with amphotericin B.
Nanoparticle carriers with miltefosine, pentamidine or SSG.
Polymeric structures like dendrimers significantly increase the solubility of hydrophobic compounds. For instance, dendrimer formulations increased amphotericin B solubility by 478-fold using an anionic linear globular dendrimer carrier [32]. Some nanoparticles have immunomodulatory properties. Stearylamine-based lipid nanoparticles without any drug increased anti-leishmanial activity by promoting a Th1 immune response, upregulating IL-12, IFN-γ, and TNF-α, and activating iNOS pathways [5]. Similarly, compounds like lactoferrin and guar gum can activate macrophages, stimulating innate immune responses [40, 76].
In addition to encapsulating approved treatments, drug repositioning remains a viable strategy for addressing leishmaniasis. Repositioned drugs, such as amphotericin B, miltefosine, and paromomycin are already used as first-line treatments in many countries.
Drug repositioning
Approximately 60% of currently approved anti-leishmanial drugs are derived from the repositioning of existing medications initially developed for diseases other than leishmaniasis [12].
The most prominent class of repositioned drugs includes antimicrobial compounds (Table 6). For instance, amphotericin B is an anti-fungal that has been successfully repositioned for visceral leishmaniasis. Other anti-fungals, such as ketoconazole, fluconazole, and itraconazole, are recommended by WHO for cutaneous leishmaniasis [110]. These drugs target the ergosterol pathway, which is essential for the cell membranes of Leishmania, offering high selectivity without affecting mammalian cells. Promising candidates like butenafine, ravuconazole, and miconazole have also demonstrated their efficacy [3, 9, 99]. Paromomycin, an antibiotic belonging to the aminoglycoside class, can be used for cutaneous leishmaniasis (CL) as a topical treatment. Clinical trials showed that paromomycin cream achieved cure rates close to 80%, providing a non-invasive and effective alternative for CL [92]. Similarly, delamanid and SQ109, originally developed for tuberculosis have shown efficacy against L. donovani. Delamanid offers the added advantage of oral administration [33, 74].
Repositioned drugs and their mechanisms of action.
Other antimicrobial repositioned drugs, such as fexinidazole and suramin, target metabolic pathways in Leishmania. Fexinidazole showed efficacy comparable to approved drugs. Indeed, 200 mg/kg of fexinidazole showed activity similar to the approved drugs miltefosine and SSG (Pentostam®). Suramin, with its immunomodulatory properties, reduced parasite burden in a mouse model infected by L. donovani [46, 74].
Anti-cancer drugs like miltefosine, ibrutinib, and imiquimod enhance host-mediated responses, particularly pro-inflammatory cytokine production [27, 103]. Miransertib and AR-12 reduced parasite burdens in vivo. Surprisingly, anti-depressants like sertraline and clomipramine have also shown potential by disrupting parasite mitochondrial functions and metabolic pathways. These drugs induced oxidative stress, ATP depletion, and apoptosis in Leishmania [52, 87].
Cholesterol reducer simvastatin boosts host defense by enhancing phagosome maturation, while rapamycin biases immune responses toward protective Th1 cytokines. The anti-arrhythmic drug amiodarone disrupts calcium homeostasis and sterol biosynthesis in Leishmania parasites [46, 71, 72].
Repositioning drugs not only leverages existing treatments, but also provides faster and more cost-effective solutions for combating leishmaniasis. However, to date, most assays using repositioned drugs against Leishmania have been conducted in vitro, with some also tested in mouse models. Many steps remain before these drugs can be used to treat patients. Combining immunomodulators with conventional treatments could further enhance their efficacy (Table 6).
Immunotherapies
Over the last 20 years, immunotherapy has been developed either as monotherapy or in combination with classically used treatments for leishmaniasis (Fig. 2). Immunity plays a critical role in the progression and outcome of the disease. Generally, Th1 immune responses involving IL-12, IL-18, IFN-g, and TNF-a are considered protective cytokines in Leishmania infections. Conversely, Th2 responses dominated by IL-4, IL-10, IL-13, and TGF-b favored parasite survival and disease progression [108]. For many years, research on leishmaniasis revolved around this Th1/Th2 dichotomy. However, it is now accepted that controlling Leishmania efficiently requires coordinated action of both pathways. Additionally, susceptibility to leishmaniasis depends on both host immunity and the virulence of the parasite species [38]. Combining current treatments with immunotherapy could reduce the concentration and duration of treatments for leishmaniasis, while shifting host immunity to favor Leishmania control.
![]() |
Figure 2 Immunochemotherapy tested in combination with drugs against Leishmania [9, 13, 35, 47, 53, 58, 63, 66, 85, 98, 107]. |
Since Th1 responses are protective against Leishmania infections, the first strategy used to enhance approved treatments has been to stimulate associated cytokines to favor remission. In dog models, domperidone and IFN-γ favor Th1 immune response and demonstrated activities against Leishmania. Domperidone is used for both prevention and treatment of canine leishmaniasis due to L. infantum [9]. Additionally, combining Glucantime® with IFN-γ has been shown to improve cure rates and reduce treatment duration, whereas Glucantime® plus active hexose correlated compound (AHCC) reduced adverse effects caused by allopurinol [9]. In mice, administering IL-12 with AMB increased the efficacy of a 2 mg/kg dose to match that of a 15 mg/kg dose [64].
Agonists of toll-like receptors (TLRs) have also been proposed. These receptors recognize pathogen-associated molecular patterns (PAMPs) [68] and trigger cytokine release via NF-κB activation, promoting pro-inflammatory responses. For example, the TLR-7 agonist imiquimod increased the efficacy of Glucantime® against L. major [98]. Similarly, the TLR-2 agonist Pam3Cys enhanced the efficacy of suboptimal doses of oral miltefosine against L. donovani in a mouse model [9].
Another strategy involves inhibiting Th2 responses by modulating cytokines. In mice, anti-IL-10R treatment accelerated L. donovani elimination and increased efficacy when combined with either Pentostam® or AMB [63–65].
In humans, the combination of IFN-γ with L-AMB has proven effective in curing a multi-resistant strain [47]. Thalidomide, an immunomodulator, reduced TNF-α secretion while increasing IFN-γ levels, thereby enhancing Th1 responses against a multi-resistant strain of mucosal leishmaniasis [35]. Despite the promising results of immunotherapies in humans, studies to date have been limited to small series or individual case reports.
Although immunotherapy is a relatively recent field, it shows promise but requires complex experimentation and development. Research into plant extracts has also emerged as a potential method to shift host immune responses favorably for Leishmania elimination.
Ethnopharmacology
Ethnopharmacology is a rapidly growing scientific field that integrates botany, chemistry, and pharmacology to evaluate the biological activity and scientific validity of plants traditionally used in medicine. By leveraging the knowledge of indigenous populations, this approach helps identify plants with potential for drug development. It can also save time by serving as a shortcut for discovering new active compounds.
All the compounds discussed in the in vivo studies (Table 7) were tested directly against Leishmania infections. These studies demonstrated reductions in parasite burden, often attributed to an increase in Th1 immune response and a decrease in Th2 cytokine secretion, thereby promoting host survival over parasite persistence. Several compounds have been specifically studied for their effects on Leishmania parasite clearance. Lupeol, mahanine, and extracts from Croton caudatus Geisel have been shown to upregulate reactive oxygen species (ROS) and nitric oxide (NO) pathways, both of which are critical for destroying intracellular pathogens [22, 25, 81]. Molecular docking identified specific parasite targets for some compounds. Lupeol, for example, bound to key Leishmania proteins such as PTR1, APRT, LPG, and GP63 [22]. Croton caudatus Geisel extract targets ascorbate peroxidase, a parasite antioxidant [25]. These finding suggested dual mechanisms, direct parasite targeting, and host immune modulation. In addition to their potential dual mechanisms, oral formulation of mahanine and JdHex offered easier administration compared to traditional anti-leishmanial drugs [25, 81].
Ethnopharmacology.
Many compounds tested in vitro demonstrated direct activity against Leishmania while others targeted the cellular pathways of host cells to control the parasite (Table 7). Some compounds inhibited essential parasite enzymes, such as arginase [55, 82, 83], or trypanothione reductase [51] or both [56]. Other plants extracts exerted their effect through cytokine modulation to enhance immune responses. For instance, Tetradenia riparia essential oil increased IFN-g secretion [24]. Modulation of phagocytic activity has also been identified as a mechanism of action including enhancement of NO secretion and phagocytic activity [79, 95] or modification in the activation state of macrophages [18]. ROS production, known to be essential in intracellular parasite clearance, was also commonly observed [18, 19, 62]. While most studies highlighted compounds that act directly against Leishmania, several reported anti-leishmanial activity without detailing the underlying mechanisms [7, 11, 97, 104], making it challenging to develop these compounds as potential treatments for Leishmania.
Discussion
This review focuses on identifying specific strategies for drug development by examining recent advances in drug discovery. The WHO currently recommended monotherapies with various approved molecules as first-line treatments, depending on the region. Among these, L-AMB is one of the most effective options; however, it requires hospitalization, is costly, and is difficult to access in many countries. Recent reports of resistant strains highlight the urgent need for innovative drug development strategies to avert a crisis akin to antibiotic resistance.
To address drug resistance, combining authorized drugs has emerged as a strategy. Combinations often reduce dosage, toxicity, and costs, while increasing efficacy. However, some combinations, such as L-AMB with SSG or miltefosine [109] or combination of intralesional Glucantime® with oral itraconazole [8] showed no benefit over monotherapy. Combining miltefosine with pentamidine resulted in additive effects, but also increased side effects and costs, rendering it impractical [93]. Developing effective combinations still requires extensive trials to optimize dosing and efficacy. While lower doses can reduce cytotoxicity and costs, these challenges remain inherent to the molecules themselves and require alternative compounds to resolve fully.
Nanocarriers, such as liposomes, have successfully reduced toxicity in AMB formulations. However, the cost increased due to reliance on animal-derived cholesterol. Using plant-derived lipids like stigmasterol (DSHemsPC) has been proposed as a promising alternative. In mice, this formulation maintained AMB levels in target organs and reduced inflammation and parasite burden [43]. Nanoparticles also enable targeted delivery, delayed release, and modified administration routes [7, 11, 97, 104]. For example, nanoparticles have facilitated oral delivery of combinations such as AMB and paromomycin [73] or topical application of AMB with miltefosine [21]. They are versatile tools for improving treatments, though they significantly increase costs.
Repositioning drugs has proven valuable in neglected tropical diseases. Repositioning saves time and money, as approved drugs have existing safety data. Miltefosine is one of the successful examples. Among repositioned drugs, only fexinidazole has undergone clinical trials for leishmaniasis. Although promising in mice, it led to relapses, causing the trial to be terminated due to insufficient efficacy [74]. However, predicting the effects of immunomodulatory drugs remains challenging. For example, simvastatin reduced parasite burden in both susceptible BALB/c and resistant C57/Bl6 mice, while pravastatin improved outcomes in susceptible mice, but worsened outcomes in resistant ones [72].
Since immunity is critical for disease control, combining immunomodulating compounds with traditional treatments has been proposed. Strategies aim to stimulate pro-inflammatory responses or reduce excessive inflammation, especially in cutaneous lesions. While some approaches have reduced treatment duration and side effects, others have shown no effect [56, 58] or increased adverse reactions as seen with pentoxifylline supplementation [13]. Combining imiquimod with Glucantime® improved cure rates in L. major infections [98], but pairing it with SSG for L. braziliensis showed no statistical significance [58]. Numerous studies have tried to increase the Th1 response by favoring a pro-inflammatory environment, but there have also been trials that inhibited Th1 responses to limit local inflammation and reduce tissue damage in response to cutaneous infection [53, 85]. Although a promising strategy, immunomodulation requires further development and cannot replace chemotherapy alone.
Ethnobotany uses local knowledge to identify plants with anti-leishmanial potential. Surveys often reveal plants with broad applications, such as treating inflammation or microbial infection [22]. However, not all studies confirm traditional uses. Some extracts exhibit high cytotoxicity, rendering them unsuitable for macrophages [30, 70]. Testing on intracellular amastigotes is critical, as Girardi et al. demonstrated that compounds active against promastigotes and axenic amastigotes failed against intracellular macrophages [34]. Few studies elucidate the mechanism of action, and effects are often attributed to synergistic interactions among multiple compounds. Despite these challenges, ethnobotany remains promising. It supports biodiversity and offers cost-effective options for natural product development. Commercializing these products aligns with growing demand for safer, environmentally conscious treatments.
Conclusion
In addition to strategies such as combining treatments, using nanoparticle formulations, repositioning drugs, employing immunomodulators, and conducting ethnobotanical research, artificial intelligence (AI) has emerged as a powerful tool in drug discovery. AI can accelerate the identification and optimization of new compounds by predicting molecular properties, optimizing chemical structures, and simulating biological interactions. Finally, AI has the potential to enhance the efficiency of drug development, while significantly reducing the time and cost of research and development.
Acknowledgments
This work was supported by the Fondation pour la Recherche Médicale, grant number ECO201806006733, to A.M.
Conflicts of interest
All the authors state that there are no competing interests.
References
- Abu Ammar A, Nasereddin A, Ereqat S, Dan-Goor M, Jaffe CL, Zussman E, Abdeen Z. 2019. Amphotericin B-loaded nanoparticles for local treatment of cutaneous leishmaniasis. Drug Delivery and Translational Research, 9, 76–84. [CrossRef] [PubMed] [Google Scholar]
- Afzal I, Sarwar HS, Sohail MF, Varikuti S, Jahan S, Akhtar S, Yasinzai M, Satoskar AR, Shahnaz G. 2019. Mannosylated thiolated paromomycin-loaded PLGA nanoparticles for the oral therapy of visceral leishmaniasis. Nanomedicine, 14, 387–406. [CrossRef] [PubMed] [Google Scholar]
- Andrade-Neto VV, Pereira TM, Canto-Cavalheiro MD, Torres-Santos EC. 2016. Imipramine alters the sterol profile in Leishmania amazonensis and increases its sensitivity to miconazole. Parasites & Vectors, 9, 183. [CrossRef] [PubMed] [Google Scholar]
- de Assis TSM, Rosa DCP, Teixeira E de M, Cota G, Azeredo-da-Silva ALF, Werneck G, Rabello A. 2017. The direct costs of treating human visceral leishmaniasis in Brazil. Revista da Sociedade Brasileira de Medicina Tropical, 50, 478–482. [CrossRef] [PubMed] [Google Scholar]
- Asthana S, Gupta PK, Jaiswal AK, Dube A, Chourasia MK. 2015. Targeted chemotherapy of visceral leishmaniasis by lactoferrin-appended amphotericin B-loaded nanoreservoir: in vitro and in vivo studies. Nanomedicine, 10, 1093–1109. [CrossRef] [PubMed] [Google Scholar]
- Asthana S, Jaiswal AK, Gupta PK, Dube A, Chourasia MK. 2015. Th-1 biased immunomodulation and synergistic antileishmanial activity of stable cationic lipid-polymer hybrid nanoparticle: biodistribution and toxicity assessment of encapsulated amphotericin B. European Journal of Pharmaceutics and Biopharmaceutics, 89, 62–73. [CrossRef] [PubMed] [Google Scholar]
- Atay I, Kirmizibekmez H, Kaiser M, Akaydin G, Yesilada E, Tasdemir D. 2016. Evaluation of in vitro antiprotozoal activity of Ajuga laxmannii and its secondary metabolites. Pharmaceutical Biology, 54, 1808–1814. [CrossRef] [PubMed] [Google Scholar]
- Bashir U, Tahir M, Anwar MI, Manzoor F. 2019. Comparison of intralesional meglumine antimonite along with oral itraconazole to intralesional meglumine antimonite in the treatment of cutaneous leishmaniasis. Pakistan Journal of Medical Sciences, 35, 1669–1673. [PubMed] [Google Scholar]
- Baxarias M, Martínez-Orellana P, Baneth G, Solano-Gallego L. 2019. Immunotherapy in clinical canine leishmaniosis: a comparative update. Research in Veterinary Science, 125, 218–226. [CrossRef] [PubMed] [Google Scholar]
- Bezerra-Souza A, Fernandez-Garcia R, Rodrigues GF, Bolas-Fernandez F, Dalastra Laurenti M, Passero LF, Lalatsa A, Serrano DR. 2019. Repurposing butenafine as an oral nanomedicine for visceral leishmaniasis. Pharmaceutics, 11, 353. [CrossRef] [PubMed] [Google Scholar]
- Bhatnagar M, Sarkar N, Gandharv N, Apang O, Singh S, Ghosal S. 2017. Evaluation of antimycobacterial, leishmanicidal and antibacterial activity of three medicinal orchids of Arunachal Pradesh, India. BMC Complementary and Alternative Medicine, 17, 379. [CrossRef] [PubMed] [Google Scholar]
- Braga SS. 2019. Multi-target drugs active against leishmaniasis: a paradigm of drug repurposing. European Journal of Medicinal Chemistry, 183, 111660. [CrossRef] [PubMed] [Google Scholar]
- Brito G, Dourado M, Guimarães LH, Meireles E, Schriefer A, Carvalho EM, Machado PRL. 2017. Oral pentoxifylline associated with pentavalent antimony: a randomized trial for cutaneous leishmaniasis. American Journal of Tropical Medicine and Hygiene, 96, 1155–1159. [CrossRef] [PubMed] [Google Scholar]
- Carvalho JDP, Assis TMD, Simões TC, Cota G. 2021. Estimating direct costs of the treatment for mucosal leishmaniasis in Brazil. Revista da Sociedade Brasileira de Medicina Tropical, 54, e04542020. [CrossRef] [PubMed] [Google Scholar]
- Casa DM, Scariot DB, Khalil NM, Nakamura CV, Mainardes RM. 2018. Bovine serum albumin nanoparticles containing amphotericin B were effective in treating murine cutaneous leishmaniasis and reduced the drug toxicity. Experimental Parasitology, 192, 12–18. [CrossRef] [PubMed] [Google Scholar]
- Chawla B, Jhingran A, Panigrahi A, Stuart KD, Madhubala R. 2011. Paromomycin affects translation and vesicle-mediated trafficking as revealed by proteomics of paromomycin -susceptible -resistant Leishmania donovani. PloS One, 6, e26660. [CrossRef] [PubMed] [Google Scholar]
- Collier MA, Peine KJ, Gautam S, Oghumu S, Varikuti S, Borteh H, Papenfuss TL, Sataoskar AR, Bachelder EM, Ainslie KM. 2016. Host-mediated Leishmania donovani treatment using AR-12 encapsulated in acetalated dextran microparticles. International Journal of Pharmaceutics, 499, 186–194. [CrossRef] [PubMed] [Google Scholar]
- Da Silva BJM, Da Silva RRP, Rodrigues APD, Farias LHS, Do Nascimento JLM, Silva EO. 2016. Physalis angulata induces death of promastigotes and amastigotes of Leishmania (Leishmania) amazonensis via the generation of reactive oxygen species. Micron, 82, 25–32. [CrossRef] [PubMed] [Google Scholar]
- Da Silva BJM, Souza-Monteiro JR, Rogez H, Crespo-López ME, Do Nascimento JLM, Silva EO. 2018. Selective effects of Euterpe oleracea (açai) on Leishmania (Leishmania) amazonensis and Leishmania infantum. Biomedicine & Pharmacotherapy, 97, 1613–1621. [CrossRef] [Google Scholar]
- Dar MJ, Din FU, Khan GM. 2018. Sodium stibogluconate loaded nano-deformable liposomes for topical treatment of leishmaniasis: macrophage as a target cell. Drug Delivery, 25, 1595–1606. [CrossRef] [PubMed] [Google Scholar]
- Dar MJ, Khalid S, McElroy CA, Satoskar AR, Khan GM. 2020. Topical treatment of cutaneous leishmaniasis with novel amphotericin B-miltefosine co-incorporated second generation ultra-deformable liposomes. International Journal of Pharmaceutics, 573, 118900. [CrossRef] [PubMed] [Google Scholar]
- Das A, Jawed JJ, Das MC, Sandhu P, De UC, Dinda B, Akhter Y, Bhattacharjee S. 2017. Antileishmanial and immunomodulatory activities of lupeol, a triterpene compound isolated from Sterculia villosa. International Journal of Antimicrobial Agents, 50, 512–522. [CrossRef] [PubMed] [Google Scholar]
- De Muylder G, Ang KKH, Chen S, Arkin MR, Engel JC, McKerrow JH. 2011. A screen against Leishmania intracellular amastigotes: comparison to a promastigote screen and identification of a host cell-specific hit. PLoS Neglected Tropical Diseases, 5, e1253. [CrossRef] [PubMed] [Google Scholar]
- Demarchi IG, Terron M de S, Thomazella MV, Mota CA, Gazim ZC, Cortez DAG, Aristides SMA, Silveira TGV, Lonardoni MVC. 2016. Antileishmanial and immunomodulatory effects of the essential oil from Tetradenia riparia (Hochstetter) Codd. Parasite Immunology, 38, 64–77. [CrossRef] [PubMed] [Google Scholar]
- Dey S, Mukherjee D, Chakraborty S, Mallick S, Dutta A, Ghosh J, Swapana N, Maiti S, Ghorai N, Singh CB, Pal C. 2015. Protective effect of Croton caudatus Geisel leaf extract against experimental visceral leishmaniasis induces proinflammatory cytokines in vitro and in vivo. Experimental Parasitology, 151–152, 84–95. [CrossRef] [PubMed] [Google Scholar]
- Diro E, Blesson S, Edwards T, Ritmeijer K, Fikre H, Admassu H, Kibret A, Ellis SJ, Bardonneau C, Zijlstra EE, Soipei P, Mutinda B, Omollo R, Kimutai R, Omwalo G, Wasunna M, Tadesse F, Alves F, Strub-Wourgaft N, Hailu A, Alexander N, Alvar J. 2019. A randomized trial of AmBisome monotherapy and AmBisome and miltefosine combination to treat visceral leishmaniasis in HIV co-infected patients in Ethiopia. PLoS Neglected Tropical Diseases, 13, e0006988. [CrossRef] [PubMed] [Google Scholar]
- El Hajj R, Bou Youness H, Lachaud L, Bastien P, Masquefa C, Bonnet P-A, El Hajj H, Khalifeh I. 2018. EAPB0503: An Imiquimod analog with potent in vitro activity against cutaneous leishmaniasis caused by Leishmania major and Leishmania tropica. PLoS Neglected Tropical Diseases, 12, e0006854. [CrossRef] [PubMed] [Google Scholar]
- Frézard F, Demicheli C, Ribeiro RR. 2009. Pentavalent antimonials: new perspectives for old drugs. Molecules, 14, 2317–2336. [CrossRef] [PubMed] [Google Scholar]
- da Gama Bitencourt JJ, Pazin WM, Ito AS, Barioni MB, de Paula Pinto C, Santos MAD, Guimarães THS, Santos MRMD, Valduga CJ. 2016. Miltefosine-loaded lipid nanoparticles: Improving miltefosine stability and reducing its hemolytic potential toward erythtocytes and its cytotoxic effect on macrophages. Biophysical Chemistry, 217, 20–31. [CrossRef] [PubMed] [Google Scholar]
- García Díaz J, Tuenter E, Escalona Arranz JC, Llauradó Maury G, Cos P, Pieters L. 2019. Antimicrobial activity of leaf extracts and isolated constituents of Croton linearis. Journal of Ethnopharmacology, 236, 250–257. [CrossRef] [PubMed] [Google Scholar]
- García-Hernández R, Manzano JI, Castanys S, Gamarro F. 2012. Leishmania donovani develops resistance to drug combinations. PLoS Neglected Tropical Diseases, 6, e1974. [CrossRef] [PubMed] [Google Scholar]
- Gaspar MM, Calado S, Pereira J, Ferronha H, Correia I, Castro H, Tomás AM, Cruz MEM. 2015. Targeted delivery of paromomycin in murine infectious diseases through association to nano lipid systems. Nanomedicine, 11, 1851–1860. [CrossRef] [PubMed] [Google Scholar]
- Gil Z, Martinez-Sotillo N, Pinto-Martinez A, Mejias F, Martinez JC, Galindo I, Oldfield E, Benaim G. 2020. SQ109 inhibits proliferation of Leishmania donovani by disruption of intracellular Ca2+ homeostasis, collapsing the mitochondrial electrochemical potential (ΔΨm) and affecting acidocalcisomes. Parasitology Research, 119, 649–657. [CrossRef] [PubMed] [Google Scholar]
- Girardi C, Fabre N, Paloque L, Ramadani AP, Benoit-Vical F, González-Aspajo G, Haddad M, Rengifo E, Jullian V. 2015. Evaluation of antiplasmodial and antileishmanial activities of herbal medicine Pseudelephantopus spiralis (Less.) Cronquist and isolated hirsutinolide-type sesquiterpenoids. Journal of Ethnopharmacology, 170, 167–174. [CrossRef] [PubMed] [Google Scholar]
- Goyonlo VM, Vahabi-Amlashi S, Taghavi F. 2019. Successful treatment by adding thalidomide to meglumine antimoniate in a case of refractory anthroponotic mucocutaneous leishmaniasis. International Journal for Parasitology: Drugs and Drug Resistance, 11, 177–179. [CrossRef] [Google Scholar]
- van Griensven J, Diro E. 2012. Visceral leishmaniasis. Infectious Disease Clinics of North America, 26, 309–322. [CrossRef] [PubMed] [Google Scholar]
- van Griensven J, Diro E. 2019. Visceral leishmaniasis: recent advances in diagnostics and treatment regimens. Infectious Disease Clinics of North America, 33, 79–99. [CrossRef] [PubMed] [Google Scholar]
- Gupta G, Oghumu S, Satoskar AR. 2013. Mechanisms of immune evasion in leishmaniasis. Advances in Applied Microbiology, 82, 155–184. [CrossRef] [PubMed] [Google Scholar]
- Gupta G, Peine KJ, Abdelhamid D, Snider H, Shelton AB, Rao L, Kotha SR, Huntsman AC, Varikuti S, Oghumu S, Naman CB, Pan L, Parinandi NL, Papenfuss TL, Kinghorn AD, Bachelder EM, Ainslie KM, Fuchs JR, Satoskar AR. 2015. A novel sterol isolated from a plant used by Mayan traditional healers is effective in treatment of visceral leishmaniasis caused by Leishmania donovani. ACS Infectious Diseases, 1, 497–506. [CrossRef] [PubMed] [Google Scholar]
- Gupta PK, Jaiswal AK, Asthana S, Verma A, Kumar V, Shukla P, Dwivedi P, Dube A, Mishra PR. 2015. Self assembled ionically sodium alginate cross-linked amphotericin B encapsulated glycol chitosan stearate nanoparticles: applicability in better chemotherapy and non-toxic delivery in visceral leishmaniasis. Pharmaceutical Research, 32, 1727–1740. [CrossRef] [PubMed] [Google Scholar]
- Heidari-Kharaji M, Taheri T, Doroud D, Habibzadeh S, Badirzadeh A, Rafati S. 2016. Enhanced paromomycin efficacy by solid lipid nanoparticle formulation against Leishmania in mice model. Parasite Immunology, 38, 599–608. [CrossRef] [PubMed] [Google Scholar]
- Hendrickx S, Van den Kerkhof M, Mabille D, Cos P, Delputte P, Maes L, Caljon G. 2017. Combined treatment of miltefosine and paromomycin delays the onset of experimental drug resistance in Leishmania infantum. PLoS Neglected Tropical Diseases, 11, e0005620. [CrossRef] [PubMed] [Google Scholar]
- Iman M, Huang Z, Alavizadeh SH, Szoka FC, Jaafari MR. 2017. Biodistribution and in vivo antileishmanial activity of 1, 2-distigmasterylhemisuccinoyl-sn-glycero-3-phosphocholine liposome-intercalated amphotericin B. Antimicrobial Agents and Chemotherapy, 61, e02525-16. [CrossRef] [PubMed] [Google Scholar]
- Kasabalis D, Chatzis MK, Apostolidis K, Xenoulis PG, Buono A, Petanides T, Leontides LS, Polizopoulou ZS, Steiner JM, Suchodolski JS, Saridomichelakis MN. 2019. Evaluation of nephrotoxicity and ototoxicity of aminosidine (paromomycin)-allopurinol combination in dogs with leishmaniosis due to Leishmania infantum: a randomized, blinded, controlled study. Experimental Parasitology, 206, 107768. [CrossRef] [PubMed] [Google Scholar]
- Khadir F, Taheri T, Habibzadeh S, Zahedifard F, Gholami E, Heidari-Kharaji M, Oryan A, Rafati S. 2019. Antileishmanial effect of rapamycin as an alternative approach to control Leishmania tropica infection. Veterinary Parasitology, 276, 108976. [CrossRef] [PubMed] [Google Scholar]
- Khanra S, Juin SK, Jawed JJ, Ghosh S, Dutta S, Nabi SA, Dash J, Dasgupta D, Majumdar S, Banerjee R. 2020. In vivo experiments demonstrate the potent antileishmanial efficacy of repurposed suramin in visceral leishmaniasis. PLoS Neglected Tropical Diseases, 14, e0008575. [CrossRef] [PubMed] [Google Scholar]
- Khodabandeh M, Rostami A, Borhani K, Gamble HR, Mohammadi M. 2019. Treatment of resistant visceral leishmaniasis with interferon gamma in combination with liposomal amphotericin B and allopurinol. Parasitology International, 72, 101934. [CrossRef] [PubMed] [Google Scholar]
- Kumar R, Sahoo GC, Pandey K, Das V, Das P. 2015. Study the effects of PLGA-PEG encapsulated amphotericin B nanoparticle drug delivery system against Leishmania donovani. Drug Delivery, 22, 383–388. [CrossRef] [PubMed] [Google Scholar]
- Kumar R, Sahoo GC, Pandey K, Das VNR, Topno RK, Ansari MY, Rana S, Das P. 2016. Development of PLGA-PEG encapsulated miltefosine based drug delivery system against visceral leishmaniasis. Materials Science & Engineering. C, Materials for Biological Applications, 59, 748–753. [CrossRef] [PubMed] [Google Scholar]
- LeishVet. A non profit scientific association. https://www.leishvet.org. [Google Scholar]
- Lima GS, Castro-Pinto DB, Machado GC, Maciel MAM, Echevarria A. 2015. Antileishmanial activity and trypanothione reductase effects of terpenes from the Amazonian species Croton cajucara Benth (Euphorbiaceae). Phytomedicine, 22, 1133–1137. [CrossRef] [PubMed] [Google Scholar]
- Lima ML, Abengózar MA, Nácher-Vázquez M, Martínez-Alcázar MP, Barbas C, Tempone AG, López-Gonzálvez Á, Rivas L. 2018. Molecular basis of the leishmanicidal activity of the antidepressant sertraline as a drug repurposing candidate. Antimicrobial Agents and Chemotherapy, 62, e01928-18. [CrossRef] [PubMed] [Google Scholar]
- Macedo SRA, de Figueiredo Nicolete LD, Ferreira ADS, de Barros NB, Nicolete R. 2015. The pentavalent antimonial therapy against experimental Leishmania amazonensis infection is more effective under the inhibition of the NF-κB pathway. International Immunopharmacology, 28, 554–559. [CrossRef] [PubMed] [Google Scholar]
- Manna L, Corso R, Galiero G, Cerrone A, Muzj P, Gravino AE. 2015. Long-term follow-up of dogs with leishmaniosis treated with meglumine antimoniate plus allopurinol versus miltefosine plus allopurinol. Parasites & Vectors, 8, 289. [CrossRef] [PubMed] [Google Scholar]
- do Carmo Maquiaveli C, Oliveira E Sá AM, Vieira PC, da Silva ER. 2016. Stachytarpheta cayennensis extract inhibits promastigote and amastigote growth in Leishmania amazonensis via parasite arginase inhibition. Journal of Ethnopharmacology, 192, 108–113. [CrossRef] [PubMed] [Google Scholar]
- do Carmo Maquiaveli C, Rochetti AL, Fukumasu H, Vieira PC, da Silva ER. 2017. Antileishmanial activity of verbascoside: Selective arginase inhibition of intracellular amastigotes of Leishmania (Leishmania) amazonensis with resistance induced by LPS plus IFN-γ. Biochemical Pharmacology, 127, 28–33. [CrossRef] [PubMed] [Google Scholar]
- Meheus F, Abuzaid AA, Baltussen R, Younis BM, Balasegaram M, Khalil EAG, Boelaert M, Musa AM. 2013. The economic burden of visceral leishmaniasis in Sudan: an assessment of provider and household costs. American Journal of Tropical Medicine and Hygiene, 89, 1146–1153. [CrossRef] [PubMed] [Google Scholar]
- Miranda-Verastegui C, Tulliano G, Gyorkos TW, Calderon W, Rahme E, Ward B, Cruz M, Llanos-Cuentas A, Matlashewski G. 2009. First-line therapy for human cutaneous leishmaniasis in Peru using the TLR7 agonist imiquimod in combination with pentavalent antimony. PLoS Neglected Tropical Diseases, 3, e491. [CrossRef] [PubMed] [Google Scholar]
- Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. 2021. Engineering precision nanoparticles for drug delivery. Nature Reviews. Drug Discovery, 20, 101–124. [CrossRef] [PubMed] [Google Scholar]
- de Morais-Teixeira E, Aguiar MG, Soares de Souza Lima B, Ferreira LAM, Rabello A. 2015. Combined suboptimal schedules of topical paromomycin, meglumine antimoniate and miltefosine to treat experimental infection caused by Leishmania (Viannia) braziliensis. Journal of Antimicrobial Chemotherapy, 70, 3283–3290. [Google Scholar]
- Moreira VR, de Jesus LCL, Soares R-EP, Silva LDM, Pinto BAS, Melo MN, Paes AM de A, Pereira SRF. 2017. Meglumine antimoniate (glucantime) causes oxidative stress-derived dna damage in BALB/c mice infected by Leishmania (Leishmania) infantum. Antimicrobial Agents and Chemotherapy, 61, e02360-16. [Google Scholar]
- Mukherjee D, Singh CB, Dey S, Mandal S, Ghosh J, Mallick S, Hussain A, Swapana N, Ross SA, Pal C. 2016. Induction of apoptosis by zerumbone isolated from Zingiber zerumbet (L.) Smith in protozoan parasite Leishmania donovani due to oxidative stress. Brazilian Journal of Infectious Diseases, 20, 48–55. [CrossRef] [Google Scholar]
- Murray HW. 2005. Interleukin 10 receptor blockade–pentavalent antimony treatment in experimental visceral leishmaniasis. Acta Tropica, 93, 295–301. [CrossRef] [PubMed] [Google Scholar]
- Murray HW, Brooks EB, DeVecchio JL, Heinzel FP. 2003. Immunoenhancement combined with amphotericin B as treatment for experimental visceral leishmaniasis. Antimicrobial Agents and Chemotherapy, 47, 2513–2517. [CrossRef] [PubMed] [Google Scholar]
- Murray HW, Flanders KC, Donaldson DD, Sypek JP, Gotwals PJ, Liu J, Ma X. 2005. Antagonizing deactivating cytokines to enhance host defense and chemotherapy in experimental visceral leishmaniasis. Infection and Immunity, 73, 3903–3911. [CrossRef] [PubMed] [Google Scholar]
- Mutiso JM, Macharia JC, Barasa M, Taracha E, Bourdichon AJ, Gicheru MM. 2011. In vitro and in vivo antileishmanial efficacy of a combination therapy of diminazene and artesunate against Leishmania donovani in BALB/c mice. Revista do Instituto de Medicina Tropical de Sao Paulo, 53, 129–132. [CrossRef] [PubMed] [Google Scholar]
- Nandan D, Zhang N, Yu Y, Schwartz B, Chen S, Kima PE, Reiner NE. 2018. Miransertib (ARQ 092), an orally-available, selective Akt inhibitor is effective against Leishmania. PloS One, 13, e0206920. [CrossRef] [PubMed] [Google Scholar]
- Netea MG, Van der Meer JWM, Sutmuller RP, Adema GJ, Kullberg B-J. 2005. From the Th1/Th2 paradigm towards a Toll-like receptor/T-helper bias. Antimicrobial Agents and Chemotherapy, 49, 3991–3996. [CrossRef] [PubMed] [Google Scholar]
- Nieto-Yañez OJ, Resendiz-Albor AA, Ruiz-Hurtado PA, Rivera-Yañez N, Rodriguez-Canales M, Rodriguez-Sosa M, Juarez-Avelar I, Rodriguez-Lopez MG, Canales-Martinez MM, Rodriguez-Monroy MA. 2017. In vivo and in vitro antileishmanial effects of methanolic extract from bark of Bursera aptera. African Journal of Traditional, Complementary, and Alternative Medicines: AJTCAM, 14, 188–197. [Google Scholar]
- Obbo CJD, Kariuki ST, Gathirwa JW, Olaho-Mukani W, Cheplogoi PK, Mwangi EM. 2019. In vitro antiplasmodial, antitrypanosomal and antileishmanial activities of selected medicinal plants from Ugandan flora: refocusing into multi-component potentials. Journal of Ethnopharmacology, 229, 127–136. [CrossRef] [PubMed] [Google Scholar]
- Oryan A, Bemani E, Bahrami S. 2018. Emerging role of amiodarone and dronedarone, as antiarrhythmic drugs, in treatment of leishmaniasis. Acta Tropica, 185, 34–41. [CrossRef] [PubMed] [Google Scholar]
- Parihar SP, Hartley M-A, Hurdayal R, Guler R, Brombacher F. 2016. Topical simvastatin as host-directed therapy against severity of cutaneous leishmaniasis in mice. Scientific Reports, 6, 33458. [CrossRef] [PubMed] [Google Scholar]
- Parvez S, Yadagiri G, Gedda MR, Singh A, Singh OP, Verma A, Sundar S, Mudavath SL. 2020. Modified solid lipid nanoparticles encapsulated with Amphotericin B and Paromomycin: an effective oral combination against experimental murine visceral leishmaniasis. Scientific Reports, 10, 12243. [CrossRef] [PubMed] [Google Scholar]
- Patterson S, Wyllie S, Norval S, Stojanovski L, Simeons FR, Auer JL, Osuna-Cabello M, Read KD, Fairlamb AH. 2016. The anti-tubercular drug delamanid as a potential oral treatment for visceral leishmaniasis. eLife, 5, e09744. [CrossRef] [PubMed] [Google Scholar]
- Ponte-Sucre A, Gamarro F, Dujardin J-C, Barrett MP, López-Vélez R, García-Hernández R, Pountain AW, Mwenechanya R, Papadopoulou B. 2017. Drug resistance and treatment failure in leishmaniasis: a 21st century challenge. PLoS Neglected Tropical Diseases, 11, e0006052. [CrossRef] [PubMed] [Google Scholar]
- Ray L, Karthik R, Srivastava V, Singh SP, Pant AB, Goyal N, Gupta KC. 2021. Efficient antileishmanial activity of amphotericin B and piperine entrapped in enteric coated guar gum nanoparticles. Drug Delivery and Translational Research, 11, 118–130. [CrossRef] [PubMed] [Google Scholar]
- Reguera RM, Morán M, Pérez-Pertejo Y, García-Estrada C, Balaña-Fouce R. 2016. Current status on prevention and treatment of canine leishmaniasis. Veterinary Parasitology, 227, 98–114. [CrossRef] [PubMed] [Google Scholar]
- Reis LES, Fortes de Brito RC, Cardoso JM de O, Mathias FAS, Aguiar Soares RDO, Carneiro CM, de Abreu Vieira PM, Ramos GS, Frézard FJG, Roatt BM, Reis AB. 2017. Mixed formulation of conventional and pegylated meglumine antimoniate-containing liposomes reduces inflammatory process and parasite burden in Leishmania infantum-infected BALB/c mice. Antimicrobial Agents and Chemotherapy, 61, e00962-17. [Google Scholar]
- Rodrigues KA da F, Amorim LV, Dias CN, Moraes DFC, Carneiro SMP, Carvalho FA de A. 2015. Syzygium cumini (L.) Skeels essential oil and its major constituent α-pinene exhibit anti-Leishmania activity through immunomodulation in vitro. Journal of Ethnopharmacology, 160, 32–40. [CrossRef] [PubMed] [Google Scholar]
- Rosenthal E, Delaunay P, Jeandel P-Y, Haas H, Pomares-Estran C, Marty P. 2009. Liposomal amphotericin B as treatment for visceral leishmaniasis in Europe, 2009. Médecine et Maladies Infectieuses, 39, 741–744. [CrossRef] [Google Scholar]
- Roy S, Dutta D, Satyavarapu EM, Yadav PK, Mandal C, Kar S, Mandal C. 2017. Mahanine exerts in vitro and in vivo antileishmanial activity by modulation of redox homeostasis. Scientific Reports, 7, 4141. [CrossRef] [PubMed] [Google Scholar]
- Santos ALS, Matteoli FP, Gonçalves DS, Seabra SH, Romanos MTV, Branquinha MH, Resende GO, Cotrim BA, Aguiar LCS, Sangenito LS. 2019. In vitro effects of the asymmetric peptidomimetic 157, containing l-tartaric acid core and valine/leucine substituents, on Leishmania amazonensis promastigotes and amastigotes. Parasitology International, 73, 101968. [CrossRef] [PubMed] [Google Scholar]
- Santos BM, Bezerra-Souza A, Aragaki S, Rodrigues E, Umehara E, Ghilardi Lago JH, Laurenti MD, Ribeiro SP, Passero LFD. 2019. Ethnopharmacology study of plants from Atlantic forest with leishmanicidal activity. Evidence-Based Complementary and Alternative Medicine: eCAM, 2019, 8780914. [CrossRef] [Google Scholar]
- Santos MF, Alexandre-Pires G, Pereira MA, Marques CS, Gomes J, Correia J, Duarte A, Gomes L, Rodrigues AV, Basso A, Reisinho A, Meireles J, Santos-Mateus D, Brito MTV, Tavares L, Santos-Gomes GM, da Fonseca IP. 2019. Meglumine antimoniate and miltefosine combined with allopurinol sustain pro-inflammatory immune environments during canine leishmaniosis treatment. Frontiers in Veterinary Science, 6, 362. [CrossRef] [PubMed] [Google Scholar]
- Schwartz J, Moreno E, Calvo A, Blanco L, Fernández-Rubio C, Sanmartín C, Nguewa P, Irache JM, Larrea E, Espuelas S. 2018. Combination of paromomycin plus human anti-TNF-α antibodies to control the local inflammatory response in BALB/mice with cutaneous leishmaniasis lesions. Journal of Dermatological Science, 92, 78–88. [CrossRef] [PubMed] [Google Scholar]
- Shalev-Benami M, Zhang Y, Rozenberg H, Nobe Y, Taoka M, Matzov D, Zimmerman E, Bashan A, Isobe T, Jaffe CL, Yonath A, Skiniotis G. 2017. Atomic resolution snapshot of Leishmania ribosome inhibition by the aminoglycoside paromomycin. Nature Communications, 8, 1589. [CrossRef] [PubMed] [Google Scholar]
- da Silva Rodrigues JH, Miranda N, Volpato H, Ueda-Nakamura T, Nakamura CV. 2019. The antidepressant clomipramine induces programmed cell death in Leishmania amazonensis through a mitochondrial pathway. Parasitology Research, 118, 977–989. [CrossRef] [PubMed] [Google Scholar]
- Singh OP, Singh B, Chakravarty J, Sundar S. 2016. Current challenges in treatment options for visceral leishmaniasis in India: a public health perspective. Infectious Diseases of Poverty, 5, 19. [CrossRef] [PubMed] [Google Scholar]
- Singh PK, Jaiswal AK, Pawar VK, Raval K, Kumar A, Bora HK, Dube A, Chourasia MK. 2018. Fabrication of 3-O-sn-phosphatidyl-l-serine anchored plga nanoparticle bearing amphotericin b for macrophage targeting. Pharmaceutical Research, 35, 60. [CrossRef] [PubMed] [Google Scholar]
- Singh-Phulgenda S, Kumar R, Dahal P, Munir A, Rashan S, Chhajed R, Naylor C, Maguire BJ, Siddiqui NA, Harriss E, Rahi M, Alves F, Sundar S, Stepniewska K, Musa A, Guerin PJ, Pandey K. 2024. Post-kala-azar dermal leishmaniasis (PKDL) drug efficacy study landscape: A systematic scoping review of clinical trials and observational studies to assess the feasibility of establishing an individual participant-level data (IPD) platform. PLoS Neglected Tropical Diseases, 18, e0011635. [CrossRef] [PubMed] [Google Scholar]
- Soltani S, Mojiri-Forushani H, Soltani S, Kahvaz MS, Foroutan M. 2020. Evaluation of antileishmanial activity employing conventional and solid lipid nanoparticles of amphotericin B on Leishmania major in vitro and in vivo. Infectious Disorders Drug Targets, 20, 822–827. [Google Scholar]
- Sosa N, Pascale JM, Jiménez AI, Norwood JA, Kreishman-Detrick M, Weina PJ, Lawrence K, McCarthy WF, Adams RC, Scott C, Ransom J, Tang D, Grogl M. 2019. Topical paromomycin for New World cutaneous leishmaniasis. PLOS Neglected Tropical Diseases, 13, e0007253. [CrossRef] [PubMed] [Google Scholar]
- Soto J, Soto P, Ajata A, Rivero D, Luque C, Tintaya C, Berman J. 2018. Miltefosine combined with intralesional pentamidine for Leishmania braziliensis cutaneous leishmaniasis in Bolivia. American Journal of Tropical Medicine and Hygiene, 99, 1153–1155. [CrossRef] [PubMed] [Google Scholar]
- Sousa-Batista A de J, Cerqueira-Coutinho C, Carmo FS do, Albernaz M de S, Santos-Oliveira R. 2019. Polycaprolactone antimony nanoparticles as drug delivery system for leishmaniasis. American Journal of Therapeutics, 26, e12–e17. [CrossRef] [PubMed] [Google Scholar]
- Souza AC, Alves MM de M, Brito LM, Oliveira LGDC, Sobrinho-Júnior EPC, Costa ICG, Freitas SDL, Rodrigues KA da F, Chaves MH, Arcanjo DDR, Carvalho FA de A. 2017. Platonia insignis Mart., a Brazilian Amazonian Plant: The stem barks extract and its main constituent lupeol exert antileishmanial effects involving macrophages activation. Evidence-Based Complementary and Alternative Medicine: eCAM, 2017, 3126458. [CrossRef] [PubMed] [Google Scholar]
- Souza RM de, Maranhão RC, Tavares ER, Filippin-Monteiro FB, Nicodemo AC, Morikawa AT, Kanashiro EHY, Amato VS. 2020. Lipid nanoparticles for amphotericin delivery in the treatment of American tegumentary leishmaniasis. Drug Delivery and Translational Research, 10, 403–412. [CrossRef] [PubMed] [Google Scholar]
- Tasdemir D, MacIntosh AJJ, Stergiou P, Kaiser M, Mansour NR, Bickle Q, Huffman MA. 2020. Antiprotozoal and antihelminthic properties of plants ingested by wild Japanese macaques (Macaca fuscata yakui) in Yakushima Island. Journal of Ethnopharmacology, 247, 112270. [CrossRef] [PubMed] [Google Scholar]
- Taslimi Y, Zahedifard F, Rafati S. 2018. Leishmaniasis and various immunotherapeutic approaches. Parasitology, 145, 497–507. [CrossRef] [PubMed] [Google Scholar]
- Teixeira de Macedo Silva S, Visbal G, Lima Prado Godinho J, Urbina JA, Souza W de, Cola Fernandes Rodrigues J. 2018. In vitro antileishmanial activity of ravuconazole, a triazole antifungal drug, as a potential treatment for leishmaniasis. Journal of Antimicrobial Chemotherapy, 73, 2360–2373. [CrossRef] [PubMed] [Google Scholar]
- Téllez J, Echeverry MC, Romero I, Guatibonza A, Santos Ramos G, Borges De Oliveira AC, Frézard F, Demicheli C. 2021. Use of liposomal nanoformulations in antileishmania therapy: challenges and perspectives. Journal of Liposome Research, 31, 169–176. [CrossRef] [PubMed] [Google Scholar]
- Tripathi P, Jaiswal AK, Dube A, Mishra PR. 2017. Hexadecylphosphocholine (Miltefosine) stabilized chitosan modified Ampholipospheres as prototype co-delivery vehicle for enhanced killing of L. donovani. International Journal of Biological Macromolecules, 105, 625–637. [CrossRef] [PubMed] [Google Scholar]
- Valle IV, Machado ME, Araújo C da CB, Cunha-Junior EF da, Silva Pacheco J da, Torres-Santos EC, Silva LCRP da, Cabral LM, Carmo FA do, Sathler PC. 2019. Oral pentamidine-loaded poly(d,l-lactic-co-glycolic) acid nanoparticles: an alternative approach for leishmaniasis treatment. Nanotechnology, 30, 455102. [CrossRef] [PubMed] [Google Scholar]
- Varikuti S, Volpedo G, Saljoughian N, Hamza OM, Halsey G, Ryan NM, Sedmak BE, Seidler GR, Papenfuss TL, Oghumu S, Satoskar AR. 2019. The potent ITK/BTK inhibitor ibrutinib is effective for the treatment of experimental visceral leishmaniasis caused by Leishmania donovani. Journal of Infectious Diseases, 219, 599–608. [CrossRef] [PubMed] [Google Scholar]
- Vásquez-Ocmín P, Cojean S, Rengifo E, Suyyagh-Albouz S, Amasifuen Guerra CA, Pomel S, Cabanillas B, Mejía K, Loiseau PM, Figadère B, Maciuk A. 2018. Antiprotozoal activity of medicinal plants used by Iquitos-Nauta road communities in Loreto (Peru). Journal of Ethnopharmacology, 210, 372–385. [CrossRef] [PubMed] [Google Scholar]
- Veeken H, Ritmeijer K, Seaman J, Davidson R. 2000. A randomized comparison of branded sodium stibogluconate and generic sodium stibogluconate for the treatment of visceral leishmaniasis under field conditions in Sudan. Tropical Medicine & International Health, 5, 312–317. [CrossRef] [PubMed] [Google Scholar]
- Velez R, Gállego M. 2020. Commercially approved vaccines for canine leishmaniosis: a review of available data on their safety and efficacy. Tropical Medicine & International Health, 25, 540–557. [CrossRef] [PubMed] [Google Scholar]
- Ventin F, Cincurá C, Machado PRL. 2018. Safety and efficacy of miltefosine monotherapy and pentoxifylline associated with pentavalent antimony in treating mucosal leishmaniasis. Expert Review of Anti-Infective Therapy, 16, 219–225. [CrossRef] [PubMed] [Google Scholar]
- Volpedo G, Pacheco-Fernandez T, Holcomb EA, Cipriano N, Cox B, Satoskar AR. 2021. Mechanisms of immunopathogenesis in cutaneous leishmaniasis and post kala-azar dermal leishmaniasis (PKDL). Frontiers in Cellular and Infection Microbiology, 11, 685296. [CrossRef] [PubMed] [Google Scholar]
- Wasunna M, Njenga S, Balasegaram M, Alexander N, Omollo R, Edwards T, Dorlo TPC, Musa B, Ali MHS, Elamin MY, Kirigi G, Juma R, Kip AE, Schoone GJ, Hailu A, Olobo J, Ellis S, Kimutai R, Wells S, Khalil EAG, Strub Wourgaft N, Alves F, Musa A. 2016. Efficacy and safety of ambisome in combination with sodium stibogluconate or miltefosine and miltefosine monotherapy for african visceral leishmaniasis: Phase II randomized trial. PLoS Neglected Tropical Diseases, 10, e0004880. [CrossRef] [PubMed] [Google Scholar]
- WHO Expert Committee on the Control of the Leishmaniases, World Health Organization. 2010. Control of the leishmaniases: report of a meeting of the WHO Expert Commitee on the Control of Leishmaniases, Geneva, 22–26 March 2010. Control de Las Leishmaniasis: Informe de Una Reunión Del Comité de Expertos de La OMS Sobre El Control de Las Leishmaniasis, Ginebra, 22 a 26 de Marzo de 2010. [Google Scholar]
- World Health Organization. https://www.who.int/news-room/fact-sheets/detail/leishmaniasis. [Google Scholar]
- World Health Organization. 2022. WHO GUIDELINE for the treatment of visceral leishmaniasis in HIV co-infected patients in East Africa and South-East Asia. [Google Scholar]
- Yang G, Choi G, No JH. 2016. Antileishmanial mechanism of diamidines involves targeting kinetoplasts. Antimicrobial Agents and Chemotherapy, 60, 6828–6836. [CrossRef] [PubMed] [Google Scholar]
- Zadeh Mehrizi T, Khamesipour A, Shafiee Ardestani M, Ebrahimi Shahmabadi H, Haji Molla Hoseini M, Mosaffa N, Ramezani A. 2019. Comparative analysis between four model nanoformulations of amphotericin B-chitosan, amphotericin B-dendrimer, betulinic acid-chitosan and betulinic acid-dendrimer for treatment of Leishmania major: real-time PCR assay plus. International Journal of Nanomedicine, 14, 7593–7607. [CrossRef] [Google Scholar]
- Zhu Y, Liao L. 2015. Applications of nanoparticles for anticancer drug delivery: a review. Journal of Nanoscience and Nanotechnology, 15, 4753–4773. [CrossRef] [PubMed] [Google Scholar]
Cite this article as: Majoor A, Michel G, Marty P, Boyer L & Pomares C. 2025. Leishmaniases: Strategies in treatment development. Parasite 32, 18. https://doi.org/10.1051/parasite/2025009.
All Tables
All Figures
![]() |
Figure 1 Current treatments and their development over time. |
In the text |
Current usage metrics show cumulative count of Article Views (full-text article views including HTML views, PDF and ePub downloads, according to the available data) and Abstracts Views on Vision4Press platform.
Data correspond to usage on the plateform after 2015. The current usage metrics is available 48-96 hours after online publication and is updated daily on week days.
Initial download of the metrics may take a while.