Previous Article in Journal
Engineering AQP1-Deficient DF-1 Suspension Cells for High-Yield IBDV Production and Vaccine Scale-Up
Previous Article in Special Issue
Biotechnology and the Future of Vaccines—From Novel Routes and Vectors to Safety, Efficacy, and Global Impact
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Overview of the Factors Related to Leishmania Vaccine Development

by
Luiz Felipe Domingues Passero
1,2,*,
Italo Novais Cavallone
1,2,3,
Gabriela Venicia Araújo Flores
1,2,3,
Sarah Santos de Lima Melchert
1,2,3 and
Márcia Dalastra Laurenti
3
1
Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil
2
Institute for Advanced Studies of Ocean (IEAMAR), São Paulo State University (UNESP), Rua João Francisco Bensdorp, 1178, São Vicente 11350-011, Brazil
3
Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School, São Paulo University, São Paulo 01246-903, Brazil
*
Author to whom correspondence should be addressed.
Vaccines 2026, 14(1), 54; https://doi.org/10.3390/vaccines14010054 (registering DOI)
Submission received: 27 November 2025 / Revised: 19 December 2025 / Accepted: 21 December 2025 / Published: 1 January 2026
(This article belongs to the Special Issue Biotechnologies Applied in Vaccine Research: 2nd Edition)

Abstract

Leishmaniasis is an infectious disease caused by several species of Leishmania parasites that preferentially infect macrophages as host cells. These intracellular parasites can evade the main microbicidal effector mechanisms of phagocytic cells and, in turn, are able to stimulate marked Th2 or regulatory T cell immune responses, which are not protective for the host. The presence of a non-protective immune response, together with the multiplication and spread of Leishmania parasites throughout the tissues, leads to the main clinical forms of leishmaniasis, such as cutaneous and visceral leishmaniasis. Although some clinical forms can be reproduced in experimental hosts such as mice and hamsters, these models do not fully mimic natural infection, which, in fact, impacts experimental vaccine development. For example, BALB/c mice are generally infected with around one million parasites, whereas humans are not infected with more than 1000 parasites, together with vector saliva. This excessive number of parasites in experimental models may affect the efficacy of vaccines in preclinical studies. Indeed, many experimental studies conducted over the past 20 years have shown only partial protection, regardless of the vaccine generation, host species employed, or the use of adjuvants. This review aims to summarize the main aspects associated with Leishmania vaccine development, including parasite diversity, host factors, immune responses, adjuvants, and antigens. Although many elegant studies have been conducted, it is possible that some essential step is still missing for the development of an effective vaccine for human use.

1. Introduction

Leishmaniasis is a group of zoonotic diseases that affect humans and several species of wild and domestic animals. It is caused by a digenetic protozoan belonging to the Trypanosomatidae family, genus Leishmania. Its biological cycle is carried out in two different hosts, one vertebrate and the other invertebrate. The vertebrate hosts include a wide variety of mammals, including rodents, edentates, marsupials, canines, and primates, including humans. Invertebrate hosts are small insects belonging to the order Diptera, family Psychodidae, subfamily Phlebotominae, and the two main genera Lutzomyia (New World) and Phlebotomus (Old World).
Leishmaniasis is endemic on five continents, in 98 countries located in tropical and subtropical regions. Currently, more than 1 billion people live in endemic areas of leishmaniasis and are at risk of contracting infection. An estimated 30,000 new cases of visceral leishmaniasis (VL) and more than 1 million new cases of cutaneous leishmaniasis (CL) occur annually [1]. Climate change, as well as the socioeconomic changes resulting from the globalization process, not only make it difficult to control the disease but also increase the number of people affected. A very striking example is the clear process of urbanization of leishmaniasis, a process closely related to rural exodus, unemployment, expansion of slums, wars, among others.
Considering the expansion of the disease, to date, there are no effective control measures such as vaccination, and the therapeutic arsenal is limited and shows, in addition to undesirable adverse reactions, reports of resistance [2]. Despite this, the development of safe and effective immune prophylactic measures should be a priority in leishmaniasis; in fact, considering all immunogens characterized and studied in several experimental models so far, why have we not yet discovered a significant and potent vaccine? To answer this question, it is very important to discuss some aspects of pathogenesis, parasite diversity, the type of vaccines and adjuvants employed so far in the context of leishmania vaccine development. Therefore, this review intends to discuss the main aspects associated with vaccine development, as well as all the significant details related to the parasite and host relationship.

2. Immune Response in Leishmaniasis

To understand the way how vaccines work in leishmaniasis, it is imperative to understand how the host immune response to Leishmania determines the fate of this parasite. The inoculation of promastigote forms by the insect vector into the vertebrate host triggers defense mechanisms related to the innate and acquired immune response.
The innate immune response begins with the action of the complement system, which has been widely studied. Antagonistic functions are attributed to the complement system: the lytic capacity of extracellular parasites and the favoring of the intracellular survival of the protozoan by aiding the phagocytosis of Leishmania sp. by macrophages, via the interaction between C3bi fragment and complement receptors found on the surface of these cells. However, the action of the complement system relies on the parasite species and on the size and composition of the lipophosphoglycan (LPG) molecule in the metacyclic parasite [3]. Furthermore, parasites can also evade the complement by the action of leishmanial protein kinase (LPK-1), which phosphorylates complement components [4].
Polymorphonuclear leukocytes are important in parasite phagocytosis. Experimental in vivo studies have shown that neutrophils are the first cells to migrate to the parasite inoculation site, resulting in parasite degeneration [4]. However, subsequent studies have shown that neutrophils would be temporary host cells for Leishmania until these infected cells undergo apoptosis. During this event, macrophages phagocytosed these infected cells without triggering the microbicidal mechanisms, allowing a silent entry of the parasites into these professional phagocytic cells, establishing infection [5].
With respect to the main antigen-presenting cells (APC), the macrophages and dendritic cells are one of the most important ones. In macrophages, the main host cell for Leishmania, the interaction between promastigote forms and the host cell involves a ligand-receptor contact, followed by a series of biochemical reactions that can lead to the activation or inhibition of the microbicidal functions of the host cell, which will determine the fate of the parasite in the host [6]. Concerning dendritic cells, studies on the role of Langerhans cells in the immunopathogenesis of cutaneous leishmaniasis caused by L. major in BALB/c mice have shown that, contrary to the assumption about the importance of these cells in the development of the cellular immune response, it has been demonstrated that dendritic cells from the dermis are the responsible for the antigen-specific stimulation of T cells [7]. Subsequent experiments showed that Langerhans cells could process and present parasitic antigens through the major histocompatibility complex II (MHCII) to CD4+ T cells, which differentiate into regulatory T cells (Treg), which will control an excessive immune response against Leishmania. Furthermore, this cell subset can produce interleukin-10 (IL-10) and tumor growth factor β (TGF-β), which directly inhibit the effector activity and proliferation of effector T cells, impacting a proper activation of macrophage, allowing the spreading and persistence of Leishmania at the site of infection [8,9]. These data suggest that Langerhans cells could inhibit inflammatory events in Leishmania infection [9]. In human localized cutaneous leishmaniasis caused by L. (Viannia) braziliensis, whose patients showed strongly positive DTH for the leishmanial antigen, the density of Langerhans cells in the inflammatory infiltrate was smaller compared to lesions caused by L. (Leishmania) amazonensis, whose patients presented less expressive or even absent DTH. The study also showed a negative correlation between the density of Langerhans cells and the density of CD4+ and CD8+ T lymphocytes in the clinical and immunopathological spectrum of American cutaneous leishmaniasis [10]. These data suggest that subsets of dendritic cells exert different immunomodulatory responses in the host during Leishmania infection. Furthermore, it has been observed that NK cells can also trigger apoptosis in infected cells, or even modulate their activity by producing IL-12 cytokine [11].
During the interaction between antigen-presenting cells and T lymphocytes, a chronic immune response will be induced and will influence the course or progression of the disease. In general, if APCs are able to differentiate T-CD4+ Th1 from naïve T cells, a high production of TNF-α and IFN-γ cytokines will be produced that would be able of activating macrophage, culminating in the elimination of the intracellular parasites [12]. In contrast, if antigen-presenting cells stimulate the differentiation of T-CD4+ Th2 cells, the parasite will reproduce, causing active lesions, since this T lymphocyte subset produces IL-4 and IL-13 cytokines, which do not activate macrophages, allowing the spread of amastigote forms. Furthermore, IL-4 aids the development of B lymphocytes, which produce and secrete antibodies, without protective effect to the host [13]. Among T lymphocytes, the presence of CD8+ T cells in the tissues infected by Leishmania sp. has been associated with healing and protection of human and murine leishmaniasis through their cytotoxic function, as well as IFN-γ production, a potent inducer of nitric oxide, which promotes the destruction of parasites [14,15].
Another subset of effector T cells is Th17, which produces IL-17 cytokine and exhibits a distinct effector function from Th1 or Th2 cells in leishmaniasis. The primary function of Th17 cells is to eliminate microorganisms through the production of IL-17, a pro-inflammatory cytokine that acts on a wide range of cell types to induce the expression of IL-6, IL-8, GM-CSF (granulocyte-macrophage colony-stimulating factor), G-CSF (granulocyte colony-stimulating factor), and metalloproteases; IL-17 is also a key cytokine to active and recruit neutrophils to the site of infection [16]. The role of Th17 cells in leishmaniasis is controversial, whereas, in cutaneous and mucocutaneous leishmaniasis, they were associated with tissue damage and pathology, but protection in visceral leishmaniasis [17,18]. Figure 1 summarizes the immune response triggered by Leishmania parasites.
Although Figure 1 summarizes the immune response of the host to Leishmania sp., it is still important to mention that differences may exist, considering the biodiversity of the hosts as well as the parasites. Furthermore, other factors, such as the saliva vector, gender, and physiological status of the hosts, can modify this prototype immune response [19,20,21].

3. Experimental Models

Experimental in vivo infection remains the optimal model for characterizing the disease and its impact on the vertebrate host. The selection of an appropriate model is predicated on two fundamental criteria: its physiological similarity to humans and its ease of handling. The most frequently utilized animal models are rodents, predominantly hamsters and mice, as they exhibit high reproductive capacity in captivity, require minimal housing and maintenance conditions, are easily handled, and are susceptible to nearly all species of Leishmania. To establish an experimental model for leishmaniasis, there are several key criteria to consider, such as genetic background of the vertebrate host, the sex of the animal, the growth phase of the parasite in culture, as well as the size of the inoculum, the route of inoculation, the site, and the method of parasite delivery. These criteria are imperative in elucidating the function of immune cells, cytokines, and effector mechanisms in regulating Leishmania parasites as well as progression or progression of the disease, as in the case of vaccination.

3.1. Experimental Models in Visceral Leishmaniasis

The dog (Canis familiaris) is considered a significant domestic reservoir of Leishmania (Leishmania) infantum, playing a key role in human infection by serving as a source of parasites to the phlebotomine sand fly vectors, which transmit VL [22]. Experimental infections using L. (L.) infantum provide a valuable laboratory model for studying VL, as they closely replicate the natural progression of the disease. In both dogs and humans, infections caused by the Leishmania (L.) donovani complex are often subclinical; however, without appropriate treatment, they can progress to severe visceral disease, potentially resulting in death. The clinical manifestations are similar in both species, typically beginning with an asymptomatic phase that advances to symptoms such as weight loss, anemia, lymphadenopathy, and fever. One notable difference is the presence of skin lesions in dogs, which are not observed in human cases [23,24,25].
Dogs are significant not only due to their epidemiological role as natural reservoirs but also to their significance as experimental models in VL research, particularly because of the similarities in disease progression between canine and human infections. In this context, Maia and collaborators reported that 75% of Beagle dogs intravenously infected with L. (L.) infantum amastigotes exhibited symptoms, including lymphadenomegaly and mild skin lesions. The spleen showed the highest parasite load, and Leishmania DNA was detected in the skin of all dogs, even in the absence of visible lesions. The authors suggest that intravenous infection with amastigotes facilitates rapid and consistent disease progression, making this model useful for evaluating therapies and vaccines. Furthermore, the detection of Leishmania in the skin of asymptomatic (experimentally infected) dogs highlights their potential role in parasite transmission to sand fly vectors [26], which was further reinforced in naturally infected dogs from endemic areas [27]. However, the use of dogs as an experimental model for VL presents several limitations, including high costs, complex maintenance, restrictions on animal numbers per experiment, and ethical considerations. Furthermore, these studies also point out some differences between the human and canine VL. Due to these constraints, rodent models are currently preferred in most experimental studies.
To overcome the issues associated with canine experimental leishmaniasis, golden hamsters and mice have been employed in the study of VL. The available results suggest that the mouse model develops a subclinical and limited infection, making this model comparable to self-controlled oligosymptomatic cases and therefore useful for studying the protective immune response [23,24]. In contrast, the experimental model of VL using the golden hamsters, Mesocricetus auratus, reproduces human disease [28]. In studies focused on viscerotropic Leishmania species, the clinicopathologic characteristics and immunopathologic mechanisms of visceral leishmaniasis (VL) in hamsters bear a striking resemblance to those observed in humans, while markedly differing from those found in murine models [28,29]. Upon systemic infection, hamsters exhibit an increased parasite load within the viscera, along with progressive cachexia, hepatosplenomegaly, pancytopenia, hypergammaglobulinemia, and, sometimes, death [30]. Although some degree of infection control is evident in the liver due to the formation of granulomas, the infection in the spleen tends to evolve progressively and chronically, leading to intense macrophagic parasitism and the disruption of the structure of white pulp with the replacement of lymphocyte cellularity by red pulp cells containing parasitized macrophages and plasma cells [31,32]. Although the hamster develops clinical-pathological characteristics and immunopathological mechanisms similar to human VL, it is important to note that it is not a perfect model; many hamsters develop ascites, renal failure, and nephrotic syndrome, conditions that are only rarely observed in humans and canine animals [29]. Nevertheless, the use of hamsters remains constrained due to a paucity of reagents (antibodies, cell markers, and cytokines) with defined specificity that are available for the study of the role of immune response in the pathology in this model.
The use of non-human primates is due to their phylogenetic proximity, anatomical and physiological similarity, and immune response resemblance to humans, making them attractive models for evaluating the pathogenicity of infectious diseases, as well as for the development of vaccines and therapies [33]. The use of the rhesus macaque (Macaca mulatta) seems to be an interesting experimental model for visceral leishmaniasis caused by L. (L.) infantum [34]. On the other hand, it was demonstrated that Cebus apella monkeys infected with L. (L.) infantum chagasi (syn. L. (L.) infantum) develop a transient infection, as these animals can mount an effective cellular immune response that controls the spread of L. (L.) infantum. According to the authors, the rhesus monkey is not a recommended model for mimicking a natural infection caused by viscerotropic Leishmania species. Non-human primates, as mentioned above, are the animals of choice for safety, immunogenicity, and protection studies in phase III clinical trials of vaccines and drug candidates for VL. However, they are more difficult to use due to high maintenance and experimental costs, strong ethical considerations, and the fact that some species are endangered. Furthermore, as observed in several studies, phylogenetic proximity does not guarantee that it is the best model for VL. Table 1 summarizes the studies that standardized models of VL.

3.2. Experimental Models in Cutaneous Leishmaniasis

The Leishmania species responsible for cutaneous leishmaniasis causes infections with different patterns of progression in animal models. In general, L. (L.) mexicana and L. (L.) amazonensis have shown high virulence in hamsters and BALB/c mice [41]. In contrast, species such as L. (V.) braziliensis and L. (V.) panamensis induce lower virulence in rodent models [42,43]. These data suggest that the differences between the evolution of infection in experimental models may limit the direct applicability of experimental data [44].
In humans, infections caused by Leishmania (L.) amazonensis exhibit a broad spectrum of clinical manifestations, ranging from localized cutaneous to mucocutaneous and diffuse cutaneous forms [45]. Infection with L. (L.) amazonensis and L. (L.) mexicana is associated with different disease patterns in mice, with some strains, such as C57BL/6 or C3H, developing chronic disease, whereas BALB/c and C57BL/10J produce progressive ulcerative lesions with cutaneous metastasis [46]. In the acute phase of L. (L.) amazonensis infection, susceptible mice (C57BL/10 and CBA) and relatively resistant mice (DBA/2) both exhibit predominant infiltration of eosinophils and mast cells. However, susceptible mice show higher numbers of amastigotes, increased tissue destruction, and a stronger humoral immune response. In resistant mice, parasite persistence is associated with infection of dendritic cells and fibroblasts [47]. The susceptibility of C57BL/10 mice to L. (L.) amazonensis is related to the absence of a Th1-type cellular immune response. This differs from BALB/c mice, where susceptibility is associated with a Th2-type response [48].
In rhesus monkeys (Macaca mulatta) infected with L. (L.) amazonensis, initial erythematous and papular skin lesions have been observed that progress to nodules that ulcerate before regressing and healing. Parasites are detected in biopsies during the initial phase of the nodular lesions. The authors suggest that the progression and resolution of L. (L.) amazonensis infection in rhesus monkeys is very similar to that observed in humans and can be used to elucidate the mechanisms of protective immunity in cutaneous leishmaniasis and to evaluate the efficacy of new vaccines [49]. Resolution has also been observed in Sapajus apella monkeys infected with L. (L.) amazonensis. However, L. (V.) braziliensis persisted with a low number of amastigote forms in the skin of S. apella primates. These results demonstrate that differences in parasite species and their immunomodulatory properties in vertebrate host cells influence the outcome of infection, as demonstrated by infection with L. (L.) amazonensis and L. (V.) braziliensis, which induce a different infection profile in the non-human primate S. apella, as observed in humans [50].
Table 2 summarizes the experimental models of cutaneous leishmaniasis standardized in recent years.
While experimental models have significantly advanced our understanding of leishmaniasis, their intrinsic limitations must also be carefully considered. Notably, no single model fully replicates the complex clinical variability observed in humans, which is shaped by factors such as host genetics, immune responses, nutrition, and co-infections. In addition, standard experimental conditions, such as high inoculum doses and the absence of sandfly saliva, differ markedly from natural transmission, potentially influencing disease progression. Furthermore, larger animal models like dogs and non-human primates present practical constraints related to high costs, logistical complexity, and ethical considerations.
In summary, animal models continue to represent valuable tools for investigating the biology of Leishmania infection and for supporting the development of effective therapies. However, careful consideration is necessary when translating experimental findings to human disease. Refinement of these models, through strategies that more closely resemble natural transmission, such as low-dose inoculation and inclusion of vector saliva, may improve their translational relevance and applicability to clinical settings.

4. Adjuvants

4.1. Description of Adjuvants

Vaccines act through the recognition of antigens by the immune system, triggering humoral and cellular responses that enhance the host’s resistance to infection [52]. Although extensive efforts have been devoted to developing vaccines against leishmaniasis, and some have been commercialized for canine visceral leishmaniasis, no vaccine is yet available for the anthroponotic form of the disease. This limitation likely arises from the inability of parasitic antigens to elicit strong cellular immunity and lasting immunological memory, which can be improved by using an appropriate adjuvant system [53].
Adjuvants are compounds of synthetic or natural origin, ranging from simple molecules to complex particulate systems, capable of functioning as immunostimulants or antigen delivery platforms [54]. They enhance immunogenicity primarily by improving antigen retention at the injection site and facilitating its uptake by immune cells. This process recruits immune cells, enables controlled antigen release, and prolongs macrophage interaction, thereby strengthening the immune response [54]. Additionally, adjuvants can activate and mature antigen-presenting cells (APCs), promote cytokine production, stimulate inflammasomes, and induce local inflammation and cell recruitment [55].
Through these mechanisms, adjuvants can reduce the required antigen dose and number of immunizations, counteract immune senescence, extend vaccine half-life, and increase antigen stability [56]. In summary, antigen–adjuvant combinations are highly effective: while antigens direct the adaptive immune response toward a specific pathogen, adjuvants activate the innate immune system via pattern recognition receptors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs) [57]. In the following sections, it will be performed a brief description of the most used adjuvants.

4.2. Alum Adjuvant

Alum forms a depot at the injection site, ensuring a slow and sustained antigen release that prolongs immune activation, enhancing antigen uptake by APCs at the depot site [50]. The inflammatory process that takes place in the dermis stimulates B lymphocytes to produce antibodies [51]. Figure 2 depicts the action mechanism of this adjuvant.
In leishmaniasis, it has been observed that alum has a limited capacity to elicit Th1 responses, restricting its applicability. Vaccines formulated with alum combined with soluble Leishmania antigen (SLA) failed to enhance protection in experimental VL. Moreover, high IL-4 levels were detected in animals receiving adjuvant alone, suggesting the intrinsic capability of alum at inducing B cell differentiation [58]. In contrast, immunization with Leishmania antigens plus Lutzomyia longipalpis peptide maxadilan (FL-MAX) combined with alum induced a more robust protection in mice, associated with lower parasite burden, reduced footpad swelling, and production of antibodies recognizing the P1, P2, and P11 regions of MAX. Nevertheless, FL-MAX caused prolonged inflammation, persisting for over 240 days post-injection [59]. Overall, alum’s efficacy in leishmaniasis is limited, as it preferentially induces a Th2 response dominated by IL-4, IL-5, and IL-10 production, an immune profile ineffective against obligate intracellular pathogens, as discussed above. However, alum’s performance improves when combined with immunostimulatory adjuvants such as BCG, which promote stronger Th1-type responses. Since alum does not activate TLRs, the inclusion of TLR agonists like BCG represents a promising strategy to enhance its immunostimulatory potential [60]; however, such a strategy makes a prototype vaccine very expensive.

4.3. Freund’s Adjuvant (FA)

Freund’s adjuvant is composed of the Complete Freund’s Adjuvant (CFA), containing heat-killed mycobacteria (Mycobacterium tuberculosis or Mycobacterium butyricum), and (2) Incomplete Freund’s Adjuvant (IFA), which lacks mycobacterial components [56]. Freund’s adjuvants generally consist of an emulsifying detergent, responsible for stabilizing antigen-containing aqueous droplets, and a mineral oil phase that slows absorption due to its hydrophobic nature. This configuration results in the gradual uptake of droplets by macrophages and prolonged antigen exposure, eliciting both Th1 (CFA) and Th2 (IFA) immune responses [61].
In CFA, the inclusion of mycobacteria introduces pathogen-associated molecular patterns (PAMPs) that activate pattern recognition receptors (PRRs), enhancing APC activity and promoting the release of pro-inflammatory cytokines such as TNF-α, IL-12, and IL-6 [62]. IL-12 further recruits NK cells, which produce IFN-γ, amplifying antigen presentation and driving Th1 differentiation [63]. Consequently, CFA induces both a depot effect and strong innate immune activation, resulting in a pronounced delayed-type hypersensitivity (DTH) response, including against self-antigens in autoimmune models [62]. By contrast, IFA, which lacks mycobacterial components, provides weaker innate activation but maintains a slow-release depot effect that favors antibody-mediated responses with a predominant Th2 profile [64,65].
In immunization protocols, CFA is commonly applied in the first dose to initiate a strong innate activation, whereas boosters employing IFA maintain immune stimulation with reduced local reactogenicity [66]. Despite their potency, CFA and IFA have limited use in humans due to adverse effects, including severe local inflammation, tissue necrosis, and potential carcinogenicity associated with the persistence of non-biodegradable mineral or paraffin oils, which may induce granulomatous lesions [67]. The detailed action mechanism of this adjuvant is illustrated in Figure 3.
In leishmaniasis, it was observed that BALB/c mice immunized three times with recombinant thiol-specific antioxidant (TSA) formulated with different adjuvants (IFA, alum + BCG, or Montanide) presented similar lymphocyte proliferation, IFN-γ production, and IgG1/IgG2a levels after challenge with L. major [68]. A similar result was obtained when IFA was formulated with total L. braziliensis antigen; however, its efficacy was similar to that of other adjuvants [69]. These data suggest that FA can be replaced by other adjuvants, considering the side effects it induces in the host. Furthermore, comparative studies already reinforced that other formulations, such as the liposomal encapsulation of antigens, exhibit higher adjuvant properties than FA [70]. Conversely, some studies reported limited vaccine efficacy in L. donovani models when the antigens lipophosphoglycan, polyacrylic acid, and a DNA vaccine (pVAX-P1) were formulated with FA, since immunized animals showed low survival (~66%), modest parasite-load reduction (38–65.8%), and failure to elicit a strong Th1 response [71,72].
Overall, the literature reveals heterogeneous results regarding CFA and IFA efficacy in experimental vaccines against Leishmania. While some studies demonstrate protective responses, others report partial or negligible efficacy, likely reflecting differences in adjuvant composition and mechanisms. CFA induces strong Th1 and Th17 responses through mycobacterial components that activate NOD2/RIP2/IRF5 pathways, generating robust IFN-γ responses essential for parasite control [62,63]. However, its intense inflammatory profile may limit consistent protection across models. In contrast, IFA, lacking mycobacteria, generally promotes weaker innate activation and a Th2-biased or mixed response, which may be less effective against leishmaniasis.

4.4. Bacillus Calmette–Guérin (BCG) Adjuvant

Bacillus Calmette–Guérin (BCG) is a live attenuated strain of Mycobacterium bovis, developed by Albert Calmette and Camille Guérin [73]. Further studies led to the development of the first oral vaccine ƒto prevent disseminated forms of tuberculosis in children, including meningitis and miliary tuberculosis. It later became one of the most widely used vaccines globally [74]. The cellular and molecular changes that BCG induces in the host were explored in Figure 4.
The use of BCG as a vaccine adjuvant against leishmaniasis has been shown to enhance pro-inflammatory cytokine production in models challenged with L. major, L. infantum, and L. donovani. BCG functions as a TLR agonist, promoting the recognition of PAMPs and, when combined with specific antigens, strengthening antigen presentation and overall immune activation, particularly through TLR2, TLR4, and TLR9 pathways [75]. In murine (Swiss albino and BALB/c mice) and hamster models of leishmaniasis, the immunization with recombinant dipeptidylcarboxypeptidase protein (rLdDCP) from L. donovani; recombinant cysteine proteinase (rLdccys1) from L. (L.) chagasi; killed L. major vaccine (KLV); autoclaved L. major (ALM) vaccine; and L. (L.) donovani antigens (triosephosphate isomerase and enolase) plus BCG adjuvant caused an increase in IFN-γ, IL-12, and NO in comparison with animals non-immunized controls and animals immunized with the antigen alone, suggesting that BCG enhanced the Th1 response and consequently reduced parasite burden in target organs of animals challenged with L. major, L. infantum, or L. donovani [76,77,78,79,80], suggesting that this adjuvant may be incorporated in experimental vaccines.

4.5. Monophosphoryl Lipid A (MPL) Adjuvant

The main natural ligand of TLR4 is lipopolysaccharide (LPS), composed of a glycan polymer, an oligosaccharide, and lipid A, the latter being essential for receptor activation due to the specific interaction with MD-2 [81]. Based on this structure, several synthetic TLR4 agonists were developed, such as monophosphoryl lipid A (MPL) and glucopyranosyl lipid A (GLA) [82]. MPL induces a systemic immune response and favors the activation of the Th1 immune response [83]. Among TLR agonists, it is the only adjuvant licensed and commercially available for use in vaccines, including the VHB vaccine Fendrix, the HPV vaccine Cervarix, and for use in patients with renal insufficiency, Fendrix vaccine being considered safe and non-toxic [84,85]. Figure 5 illustrates the action mechanism of this adjuvant.
In a comparative study, Thakur et al. assessed the prophylactic effect of killed L. donovani antigens (KLD) formulated with alum, saponin, cationic liposomes, or MPL in BALB/c mice. Although killed antigen preparations typically show limited efficacy [86], the addition of cationic liposomes or MPL substantially enhanced protection to 83.4–92.8% and 86–93.7%, respectively, and enhanced a Th1 immune response [86]. A similar response was observed when hamsters were immunized with the recombinant antigens rA6 or rF14 in the presence of MPL adjuvant in visceral leishmaniasis [87].
The recombinant polyprotein rLeish-111f, composed of TSA, LmSTI1, and LeIF antigens, was formulated with MPL-SE. Following three subcutaneous immunizations, mice developed mixed IgG1/IgG2a responses, high IFN-γ levels, and protection to L. major challenge comparable or superior to soluble leishmanial lysate [88]. Subsequently, Fujiwara et al. formulated the same experimental vaccine in the presence of MPL-SE or AdjuPrime that was administered subcutaneously in three doses at four-week intervals in dogs. Both adjuvants induced high IgG2 titers, but the vaccine built with MPL-SE triggered a markedly stronger Th1 response, with an IgG2/IgG1 ratio ~40-fold higher than AdjuPrime, suggesting superior protective potential against canine visceral leishmaniasis [89]. However, MPL-SE failed to provide efficacy in a phase III trial of the Leish-111f vaccine in dogs challenged with L. infantum, where more than 87% of vaccinated animals developed disease [90].
In humans with mucocutaneous leishmaniasis receiving sodium stibogluconate treatment, adjunct immunization with LEISH-F1 formulated in MPL-SE (three doses at 28-day intervals) was safe, well-tolerated, and immunogenic. Vaccination increased LEISH-F1-specific IL-2-producing memory CD4+ T cells, a response associated with clinical cure, supporting the safety and immunogenicity of LEISH-F1/MPL-SE in patients [91].

4.6. CpG-Oligodeoxynucleotides (CpG ODN) Adjuvant

CpG ODNs are short synthetic single-stranded DNA molecules that mimic bacterial DNA and function as immunomodulatory adjuvants by activating TLR9 within endocytic vesicles of plasmacytoid dendritic cells (pDCs) and B cells, thereby initiating innate and adaptive immune responses [92], resulting in the recruitment of macrophages, NK cells, and T lymphocytes to the immunization site [93].
Despite their overall Th1-polarizing activity, CpG ODNs encompass at least three classes with distinct immunological profiles [94]. Type B (K) CpGs primarily activate B cells and antigen-presenting cells, inducing IL-6 and IL-12 production and stimulating Th1 differentiation with robust IgG2a and IgM responses but minimal IFN-α and IFN-γ induction [93]. Type A CpGs, in contrast, strongly induce IFN-α secretion and promote the maturation of APCs and pDCs [95]. Type C CpGs combine features of both classes, simultaneously stimulating IL-6 production by B cells and IFN-α secretion by pDCs [93]. The molecular and cellular mechanisms that CpG induces in the host are illustrated in Figure 6.
Different experimental approaches have evaluated CpG ODN as an adjuvant in Leishmania vaccine formulations. When combined with soluble L. major antigen (SLA), CpG ODN markedly increased anti-Leishmania IgG2a and IFN-γ levels in BALB/c mice compared with SLA alone [96]. Subsequent studies demonstrated that incorporating CpG ODN into delivery systems further strengthened its immunostimulatory effect. Tafaghodi and colleagues investigated CpG ODN in combination with autoclaved L. major (ALM) antigens encapsulated in poly(d,l-lactide-co-glycolide) (PLGA) nanospheres or alginate (ALG) microspheres. In both systems, BALB/c mice receiving ALM + CpG ODN, either free or encapsulated, exhibited higher IgG2a/IgG1 ratios, increased IFN-γ production, and reduced IL-4 levels relative to ALM alone or infected controls [97]. These findings confirmed that CpG ODN promotes a Th1-biased response and that encapsulation strategies can prolong and amplify this protection.
The adjuvant effect of CpG ODN was also validated using recombinant antigens. Its association with SODB1 and Pxn4 reduced lesion size and parasite burden in BALB/c mice challenged with L. donovani, outperforming recombinant antigens alone [98]. Likewise, CpG ODN formulated with recombinant L. major STI1 (rLmSTI1), either free or in liposomes, induced smaller lesions, lower splenic parasite loads, and higher IgG2a responses compared with non-adjuvanted groups, consistent with a CpG-driven Th1 profile [99].
Overall, these studies highlight CpG ODN as a promising adjuvant capable of eliciting strong and protective Th1-type immunity in Leishmania models. However, its activity is species-dependent, robust in mice due to broader TLR9 expression, but more restricted in humans, where TLR9 is limited to plasmacytoid dendritic cells and select B-cell subsets [100]. Thus, large-animal models may provide an essential translational bridge for advancing CpG-based vaccine formulations toward clinical development [101].
Adjuvants play an essential role in amplifying immune responses to antigens; however, conventional adjuvants often display limited immunostimulatory capacity, underscoring the need for next-generation adjuvants. An ideal adjuvant should be safe, stable before administration, biodegradable, and easily eliminated while promoting a targeted and effective immune response and remaining cost-effective for large-scale production. Advancing research on the mechanisms and immunological properties of various adjuvants is fundamental for rational vaccine design, enabling the development of more effective and personalized immunization strategies.

5. Vaccines for Leishmaniasis—20 Years of Studies on the First, Second, and Third-Generation Vaccines

Studies of vaccination employ different platforms to produce vaccines, in leishmaniasis some of them have pointed out the significance of second- and third-generation vaccines in inducing effective and long-lasting immunity over live or first-generation vaccines. In fact, the COVID-2019 pandemic also reinforced this statement, by showing the World an important breakthrough in human medical history, which a global vaccination with mRNA vaccines, viral vectors, or protein subunit vaccines avoided around 20 million deaths [102]. In fact, the global emergence was huge, allowing a fast and reliable process of vaccine production given the availability of the virus strains, viral genome, and identification of the conserved spike protein. Furthermore, mRNA vaccines, non-replicating viral vectors, and lipid nanoparticle systems was already recognized as a safe strategy to be employed in different medical purposes. Although pandemics brought irreparable losses worldwide, it also gave us significant lessons: (1) global collaboration in science is essential; (2) it is imperative to obtain as much as possible information about the genome and relationship of infectious agents and hosts; (3) different vaccine platforms allow a rapid development of vaccines and (4) vaccines are safe to humans. Unfortunately, in neglected diseases, although substantial efforts have been made by the scientists, few candidates have been approved to be translated into human health, as is the case of the vaccine RTS,S/AS01 (Mosquirix) and R21/Matrix-M, both approved by the World Health Organization and used to protect humans from malaria [103]. The vaccines cited above are the first ones approved in the context of parasitic infections, even inducing partial protection. In fact, this experience with malaria vaccine brought the idea that it is possible to build and develop a vaccine for neglected diseases even in the context of parasites with complex life cycles and different interactions with humans, vectors, wild or peri-domestic animals, as is the case with leishmaniasis. In this specific situation, this task should be significantly difficult due to the existence of a diversity of parasite species causing a spectrum of human diseases, which in turn, makes it hard to obtain a single vaccine able to confer protection across most pathogenic species. The following sections will discuss some vaccine platforms and the main findings on the experimental vaccines developed in the last 20 years. Articles were selected after a bibliographic survey using a Boolean search, performed in the Scopus and PubMed databases, during November 2024 to March 2025 and the combination of words was used to expand the possibility of finding data that would meet the expectations of the present study: “(vaccine OR vaccination OR immunization OR leishmanization)” AND (first-generation vaccine OR whole cell lysate OR total antigen) AND (second-generation vaccine OR subunit vaccine OR protein) AND (third-generation vaccine * OR DNA vaccine OR RNA vaccine)”.

5.1. Leishmanization and the Generations of Vaccines Against Leishmaniasis

In the context of vaccination, some authors consider live vaccines as the first generation [104]; we have chosen to include them in the topic of leishmanization, discussed in the next paragraph.
Leishmanization refers to the first prophylactic attempts against leishmaniasis, which involve the inoculation of live parasites in less exposed areas of the body to induce immunity against more severe subsequent infections [105]. Studies on leishmanization began in 1940 and continued until the 1980s, using strains of L. tropica [98,106,107] and L. major [108]. Despite demonstrating protection against reinfections in humans, issues such as parasite persistence, long-lasting and non-self-healing wounds, need for treatment in some cases, hypersensitivity reactions, and lack of standardization were reported [98,106,107,108], preventing further development and prompting the search for safer vaccines [109].
During the past 20 years, some studies have maintained the principles of leishmanization while modifying the method to produce a safer vaccine. In this method, Laabs and colleagues [110] immunized C57BL/6 intradermally with 104 live L. major promastigote forms plus CpG DNA adjuvant. This immunization protocol triggered IL-2 production by dendritic cells and IFN-γ by NK cells. Furthermore, vaccinated animals did not develop skin or mucosal lesions; however, no subsequent challenge was performed [110]. In an earlier study by the same group [111], it was observed that 100% of mice vaccinated exhibited protection against a subsequent challenge (15 weeks after vaccination) with 500 metacyclic promastigotes (an estimated concentration of a natural sandfly inoculation) [111].
Leishmanization studies employing L. tarentolae, a non-pathogenic species for humans, have been reported [112,113,114]. BALB/c mice were vaccinated intraperitoneally with 5 × 106 stationary-phase promastigotes of L. tarentolae in a single dose, and after 6 weeks post-vaccination, these animals exhibited cross-protection after challenge with 5 × 107 promastigotes of L. donovani, since these animals exhibited 80% less parasites in the spleen and liver than non-vaccinated mice [113]. The immunogenic and protective potentials of L. tarentolae were highlighted in another study, in which BALB/c mice were vaccinated intraperitoneally with 5 × 106 recombinant L. tarentolae expressing the A2 gene of L. donovani (a virulence factor expressed in amastigotes of virulent Leishmania strains) in a single dose [112]. Six weeks after immunization, animals received an inoculum of 107 L. infantum intravenously. Vaccinated animals exhibited 90% less parasites in the spleen and liver, which was associated with the elevation of IFN-γ levels and reduction in IL-5, suggesting that this vaccine protects animals by increasing the Th1 immune response [112]. A more recent study demonstrated that the immunization with a single dose containing 2 × 105 live L. tarentolae promastigotes with CpG ODN protected BALB/c mice from the challenge with 2 × 105 L. major metacyclic promastigotes [114].
Another approach consists of the use of live attenuated strains of Leishmania. The attainment of the result can be achieved in several ways, such as culture in media supplemented with antibiotics such as gentamicin [115], genetic modifications for drug susceptibility (e.g., Ganciclovir and 5-fluorocytosine) [116], deletion of several genes such as HSP70-II [117] and centrin [109,118,119]. Furthermore, the use of naturally attenuated strains such as Sri Lankan L. donovani, which does not cause visceral disease [120], is another interesting possibility. A study that followed dogs for 24 months after a subcutaneous single-dose vaccination with 5 × 107 gentamicin-attenuated L. infantum stationary-phase promastigotes demonstrated that 100% of the vaccinated group showed no leishmanial DNA, and 97.8% of animals exhibited no clinical signs of the disease [115], this protection was associated with higher levels of IgG2 compared to the IgG1 levels found in the sera of vaccinated animals, as IgG2 antibodies are associated with asymptomatic infection, whereas IgG1 is related to disease progression [115].
Davoudi and collaborators studied the protective potential of subcutaneous leishmanization in C57BL/6 mice using 2 × 106 promastigotes of L. major (Lmtkcd+/+), which were susceptible to Ganciclovir and 5-fluorocytosine due to the inclusion of thymidine kinase (tk) and cytosine deaminase (cd) genes in the genome of this parasite, respectively [116]. The enzyme encoded by the tk gene targets ganciclovir, a nucleoside analog that blocks DNA replication, while the enzyme encoded by the cd gene acts on the 5-fluorocytosine molecule, generating the active drug metabolite that inhibits RNA synthesis [121]. The mice were treated with both drugs on day 8 post-vaccination to attenuate the drug-sensitive strain and, after 3 weeks, C57BL/6 mice were challenged with wild-type L. major promastigotes; studies on lesion size and tissue parasitism showed that vaccinated animals did not develop lesions and that the parasite burden in the spleen was reduced by 90% in comparison to the non-vaccinated mice [116]. Another study showed that the leishmanization in BALB/c mice employing HSP70-II knockout L. infantum promastigotes provided more than 90% cross-protection against an infection with 103 L. major metacyclic promastigotes, as measured by the splenic parasite load [117]. In another study, it was observed that intradermal vaccination with centrin-deleted L. major stationary-phase promastigotes (Lmcen−/−) offered 90% reduction in the spleen and liver parasite loads in hamsters subsequently infected (seven weeks later) with L. donovani promastigotes or exposed to infected sandflies [118].
Leishmanization represents the historical foundation for the development of vaccines against leishmaniasis, and current studies employing this methodology in murine and canine models have enriched the literature regarding the understanding of protective immunity against the parasites. It is notable that leishmanization using non-pathogenic, attenuated strains or those associated with Th1 immune-stimulating adjuvants has achieved remarkable results in reducing parasite burden and providing protection against infection challenges. However, the parasite is not completely neutralized or eliminated, rendering these vaccines neither effective nor safe. The demand for a more humanized immunization approach and the reported evidence of cross-immunity (the ability of immunization with one strain to confer protection against infection by different Leishmania strains) have encouraged investigations into the subsequent three generations of vaccines. Table 3 summarizes the main data on leishmanization.

5.2. First Generation of Leishmania Vaccines

The first generation of vaccines consists of killed parasite antigens, which can be obtained by different chemical methods, such as the use of formalin and merthiolate [123,124], or physical methods, such as sonication, autoclaving, freezing and thawing, and pasteurization [125]. This class of vaccines does not bring significant concerns regarding the emergence of clinical signs and symptoms, transforming the vaccinated individual into a reservoir, allowing the transmission of parasites by a vector or even a possible reactivation of the pathogenic state of the attenuated Leishmania strains used in protocols of leishmanization [126,127].
Current studies with first-generation vaccines use various methods for obtaining total antigens, different adjuvants, and different hosts, such as rodents [123,124,125,128], dogs [129,130,131], and humans [132]. Experimentally, a vaccine produced with the whole cell lysate of L. amazonensis promastigote forms was tested with different preservative agents, such as thimerosal and phenol [123]. Two groups of C57BL/10 mice were vaccinated with these vaccines (100 mg/dose) plus Corynebacterium parvum as adjuvant (250 mg/dose) in two subcutaneous doses. This adjuvant was used due to its ability to activate both cellular and humoral immune responses [133]. Seven days after the last immunization, the animals were challenged with L. amazonensis promastigotes. Both vaccines conferred 50% protection (defined by the appearance of lesions); however, animals immunized with the phenol-preserved vaccine developed smaller wounds than those seen in animals immunized with the thimerosal-preserved vaccine; in addition, among vaccinated animals that developed lesions, they were ten times smaller compared to those observed in control groups [123].
Germanó and collaborators (2020) evaluated different methodologies to obtain L. amazonensis first-generation vaccines, such as autoclaving, freeze-thawing, pasteurization, and sonication [125]. These experimental vaccines were combined with the adjuvant polyinosinic:polycytidylic acid [(Poly (I:C)], a TLR3 agonist [125]. Once formulated, BALB/c mice were immunized with three doses of these vaccines (100 µg/dose of L. (L.) amazonensis extracts plus 50 µg of Poly (I:C) by the subcutaneous route, animals were challenged with L. (L.) amazonensis promastigotes [125]. The group of animals vaccinated with the sonicated formulation associated with poly (I:C) adjuvant exhibited the smallest lesion size as well as less splenic involvement. Furthermore, such animals produced the highest levels of anti-Leishmania IgG1 and IgG2a isotype in comparison to the other first-generation vaccines, it was observed that the IgG2a/IgG1 ratio was higher [125]. This vaccine formulation stimulated a classical Th1-type response, but the increase in IL-4 levels and the production of basal levels of IL-10 may have contributed to the persistence of parasites in vaccinated animals [125].
The immunogenic and protective potential of L. (V.) shawi amastigote and promastigote forms was also analyzed as first-generation vaccines [134]. BALB/c mice received the whole cell lysates of amastigote or promastigote forms (25 μg/dose) subcutaneously twice at a regular interval of seven days. After the challenge with L. shawi promastigotes, it was observed that animals immunized with promastigote whole cell lysate developed smaller lesions and exhibited 70% less skin parasitism compared with non-immunized animals. The partial protection was associated with the elevation of IFN-γ, IL-12, and TGF-β cytokines. In contrast, immunization with amastigote forms whole cell lysate exacerbated the infection, increasing lesion size by 50% and skin parasite burden by 450% compared with non-immunized animals, this response occurred due to the high levels of TGF-β and low of IL-2, IFN-γ, and nitric oxide produced by mononuclear cells [134]. These findings highlight that antigens derived from amastigote forms show immunosuppressive properties, while those from promastigote forms have an immunostimulatory potential in the host.
First-generation vaccines have already been studied in combination with saponin adjuvant [129,130] and saliva gland extract from Leishmania vectors. In this sense, two experimental vaccines were formulated, the first one with the whole antigen of L. (V.) braziliensis (LbSap) [129] plus saponin, and another constituted with L. (V.) braziliensis whole antigen plus salivary gland extract (SGE) of Lutzomyia longipalpis and saponin (LbSapSal) [130]. These vaccines were administered subcutaneously to dogs in a three-dose regimen at four-week intervals, each dose [129,130]. Both vaccines induced a cross humoral immune response with anti-Leishmania chagasi antibodies IgG1 and IgG2, while vaccination with LbSapSal also developed anti-SGE IgG1 and IgG2; both immunizations stimulated the proliferation of B, T CD4+ and CD8+ T lymphocytes [129,130]. Furthermore, it was observed that increased CD4+ and CD8+ T cell populations were correlated with asymptomatic animals showing low parasite burden [129,130,135]. Both vaccines stimulated MHC-I expression, and in in vitro experiments using PBMCs showed elevated NO production in the supernatants of cells from vaccinated animals stimulated with soluble antigens of L. braziliensis and L. chagasi [129,130]. Thus, these vaccines exhibited strong antigenic potential and could activate an immune response associated with the resistance pole; however, further studies on cytokine profiling are necessary to better understand the T cell response elicited by this type of immunogen in dogs [129,130].
A formalin-killed L. major promastigotes vaccine (FKP) was formulated with several adjuvants, such as Montanide ISA 720 [136], Bacille Calmette–Guerin (BCG), and Alum [124]. BALB/c mice were subcutaneously vaccinated with three doses of this vaccine and then challenged with L. major promastigote forms [124]. Among all vaccines tested, the one formulated with the Montanide ISA 720 adjuvant generated the highest IFN-γ production, the lowest parasite load (96% reduction), and the smallest lesion size (80% reduction) compared to the control animals. In contrast, animals vaccinated with FKP without any adjuvant exhibited intermediate lesion sizes, lower IFN-γ production, and a higher parasite burden compared to the other vaccinated groups [124]. These studies highlight the importance of using adjuvants to provide greater protection in different experimental models.
The combination of antigenic extracts from different species of Leishmania has also been reported during the last 20 years of vaccine research [131]. Giunchetti and collaborators developed a vaccine containing L. amazonensis and L. braziliensis whole parasite lysates plus BCG as an adjuvant. This vaccine was assessed in beagle dogs by administering three subcutaneous doses at four-week intervals. It was observed that vaccination employing L. (L.) amazonensis and L. (V.) braziliensis whole parasite lysates, especially after the second and third immunizations, caused a significant increase in IgG anti-Leishmania antibodies. Furthermore, it was observed that these antibodies could recognize L. chagasi antigens [131], suggesting that this experimental vaccine would be able to induce cross-protection. Subsequently, Grenfell et al. developed two vaccines, one containing crude extracts of L. braziliensis promastigotes plus saponin (50 µg) and the second one was constituted by L. amazonensis promastigotes and saponin; both vaccines were administered in groups of BALB/c mice in three subcutaneous doses at one-week intervals [128]. In this study, the main objective was to investigate whether such vaccines were able to confer cross-protection after challenge with L. chagasi promastigotes. In fact, after challenge with infective L. chagasi, the immunized animals presented a reduction of 25% and 50% in the hepatic and splenic parasite loads, respectively. Furthermore, a significant inhibition of IL-4 and IL-10 production was observed [128]. The reactivity of anti-L. amazonensis antibodies to L. chagasi antigens indicate an analogous antigenic repertoire between these species, allowing for an increased spectrum of vaccine protection by using these species combined in the vaccine composition [131].
Some studies have also been conducted in humans. Vélez and collaborators [132] assessed the cross-immunity of the Leishvacin®, composed of merthiolate-killed L. amazonensis in humans [132]. Three intramuscular doses (0.36 mg/mL/dose) were administered at 20-day intervals to a group of soldiers who worked in endemic areas of L. panamensis infection. At the end of 15 months of monitoring, it was observed that the incidence of cutaneous leishmaniasis in vaccinated and placebo groups was similar (7.8% and 6.8%, respectively), concluding that the vaccine, despite being safe, did not provide protection against L. panamensis [132]. Khalil and collaborators demonstrated that a single vaccination using killed L. major plus BCG in children stimulated a cellular immune response in 50% of the patients, as measured by the skin test [137]; however, the immune response decreased after 18 months of the immunization to 25%, suggesting that this immunization scheme does not induce a long-lasting immunity [137]. Possibly, the failure of this vaccination is associated with the presence of several antigens able to induce and expand Th2 lymphocyte clones, restricting the potential of first-generation vaccines [134].
Although these studies show some discrepant results on the protective effect of first-generation vaccines during experimental and natural infections, all these studies pointed out the safety profile of such vaccines in different hosts, which in turn suggests that defined antigens will also present a safe profile during immunizations. Table 4 summarizes the main findings about first-generation vaccines.

5.3. Second Generation of Leishmania Vaccines

Studies with first-generation vaccines allow the investigation of safer prophylactic methods than leishmanization. However, it is evident that although the total antigenic extract provides protection against subsequent challenge, not all antigens are responsible for this protection [143]. This observation enabled the development of the second generation of vaccines, which involves the isolation of a specific parasite antigen; the modification of proteins and the production of chimeras; and the use of the secretome produced by Leishmania in vaccination protocols [104].
Among the defined Leishmania antigens studied over the past 20 years are the Leishmania-Activated C-Kinase Antigen (LACK) [144,145]; histones [146]; hydrophilic acylated surface protein B (HASPB) [146]; the 63-kDa metalloprotease (GP63) [147]; among others. The LACK antigen, also known as the Leishmania homologue of receptors for activated C kinase or P36 (a 36-kDa protein), is conserved in Leishmania species and is expressed in both promastigote and amastigote forms [148], features that make this antigen a significant protein to be studied in immunization protocols. Stober and collaborators [144] studied the antigenic potential of a recombinant L. major’s LACK protein that was expressed in Escherichia coli. BALB/c mice were vaccinated subcutaneously with two doses of 100 µg of LACK, followed by a challenge with L. major promastigote [144]. It was verified that immunized animals produced high levels of IFN-γ; however, the LACK vaccine failed to protect against infection, possibly due to increased IL-10 production [144]. More recently, it was demonstrated that LACK from L. donovani in the recombinant form also fails to induce protection in visceral experimental leishmaniasis [144]. Although LACK is a conserved antigen among Leishmania species and is expressed in all stages of the parasite, it has no antigenic ability to induce a protective immune response, thus encouraging the search for new antigenic factors for vaccine development.
GP63, or leishmanolysin, is a glycoprotein abundantly expressed on the plasma membrane of promastigotes and at lower density on the membrane of Leishmania amastigotes [149]. It is a zinc-dependent metalloprotease, which enables it to cleave host proteins and thus evade the host immune response [149]. This antigen has been studied extensively in the host and parasite context, including the development of vaccines. The studies conducted by Kaur et al. focused on the formulation of vaccines containing GP63 alone (10 µg/dose) or in combination with other antigens such as heat shock protein 70 kDa (Hsp70, 10 µg/dose), total autoclaved L. donovani antigens (ALD, 2.5 mg/dose), or the adjuvant monophosphoryl lipid A (MPL-A) [150,151]. BALB/c mice were vaccinated with two subcutaneous doses of these vaccines; the animals were challenged intracardially with L. donovani promastigotes [150,151]. All vaccine formulations resulted in progressive reductions in parasite burden in the spleen and liver at the analyzed experimental time points, with the formulation GP63 + Hsp70 + MPLA providing the highest level of protection. In addition, all formulations induced the production of Th1-type cytokines (IFN-γ and IL-2) and reduced the levels of Th2 and regulatory cytokines (IL-4 and Treg). However, mice immunized with the GP63 + Hsp70 + MPLA vaccine displayed the most effective microbicidal cytokine response among all formulations. Control formulations lacking GP63 failed to induce antigenic or protective responses, highlighting the importance of developing vaccines that include the GP63 antigen.
Another study showed that a vaccine containing 2.5 µg of gp63, purified from L. donovani promastigotes and encapsulated in cationic liposomes, was administered intraperitoneally in three doses to BALB/c mice, which were subsequently challenged with L. donovani promastigotes [147]. All immunized mice survived up to three months after challenge, while 50% of non-immunized mice died; additionally, the vaccine reduced spleen and liver parasite loads by 80% compared to unvaccinated animals. Furthermore, it was observed that these vaccine formulations induced proliferation of CD4+ and CD8+ T cell along with an elevation in IFN-γ production, accounting for the partial protection observed. These studies identify GP63 as an immunogenic and protective component against infection, effective across different inoculum sizes and various formulations and delivery systems (both free and liposomal forms).
Histones are highly conserved proteins that associate with DNA to structure chromatin. Like other eukaryotic organisms, histones are present in Leishmania protozoa with a high degree of conservation among species [104]. HASPB is a membrane protein found in the extracellular and intracellular forms of Leishmania sp.; therefore, it can be a significant target to develop efficient prophylactic measures. The protective immune response potential of L. infantum histone and HASPB recombinant proteins was studied separately and in combination [146]. Three vaccine formulations were produced with the recombinant proteins: H1 (100 µg of histone 1), HASPB1 (100 µg of HASPB1), and H1+HASPB1 (100 µg of each protein); Montanide ISA 720 adjuvant was combined with such vaccines. The dogs were vaccinated with three intradermal doses of each formulation, and 45 days after the last immunization, the animals were challenged with L. infantum promastigotes. Adverse reactions were not recorded during immunizations, except for local inflammation after the second dose [146]. Following challenge, protection against the manifestation of symptoms (lymphoadenopathy, onychogryphosis, alopecia, cutaneous lesions, weight loss, or keratoconjunctivitis) was observed: HASPB1 and H1+HASPB1 protected 50% of dogs from developing leishmaniasis symptoms, while the H1 vaccine protected 62.5% of the animals, which also had the lowest parasite load in the bone marrow and lymph nodes [146]. It is important to emphasize that this protection was observed under extreme experimental conditions, in which the infection inoculum was overestimated (108 parasites). The vaccines exhibited a partial protective effect in dogs, and it was observed that the vaccine composed exclusively of the H1 protein was more effective than the combination of antigens. This finding highlights the importance of antigen selection in vaccine design, as combining antigens may impair the overall immune effect.
The antigenic potential of other L. donovani proteins has been investigated and classified according to their molecular weights: 91 (LD91), 72 (LD72), 51 (LD51), and 31 (LD31) kDa [152] as well as a recombinant protein with 78 kDa [153]. Proteins extracted from stationary-phase L. donovani promastigotes were incorporated into cationic liposomes, generating four vaccines containing 2.5 µg of their respective antigens (LD91, LD72, LD51, LD31). These vaccines were administered intraperitoneally in three doses to BALB/c mice; ten days after the final dose, the animals were challenged with 2 × 107 stationary-phase L. donovani promastigotes. The LD91 antigen provided low protection after challenge, whereas the LD72, LD51, and LD31 proteins reduced parasite loads in these organs by 60–70%. Two additional vaccines were formulated using the recombinant protein with 78 kDa: (1) 10 µg of r78 + 40 µg of MPL-A, and (2) 10 µg of r78 encapsulated in cationic liposomes. The vaccination protocol consisted of three intraperitoneal injections to BALB/c mice; fifteen days after the last dose, the animals were challenged with L. donovani promastigotes. The r78 protein conferred a progressive reduction in parasite burden in infected animals—from 47% (r78 + MPL-A) and 49% (r78 in liposomes) at 15 days post-challenge to 96% and 97%, respectively, at 90 days post-challenge. In addition, both r78-based vaccines stimulated IFN-γ production throughout the experimental period.
Other proteins have been studied in recent years. Vakili and colleagues produced a multi-epitope vaccine composed of four antigenic proteins: histone H1, sterol 24-C-methyltransferase (SMT), a hypothetical protein specific to Leishmania (LiHy), and an antigenic protein specific to Leishmania (LSAP) [154]. This vaccine, tested alone (30 μg of the protein) or combined with Freund’s adjuvant [30 μg of the protein in 30% (v/v)], was administered in two subcutaneous doses (2-week intervals) to BALB/c mice, followed by the challenge with L. infantum promastigotes. Both formulations stimulated IFN-γ production and inhibited IL-10 production, culminating in a significant reduction in the parasite load of the spleen by 80%. Lage and colleagues developed a vaccine using a protein they termed ChimeraT, produced with prohibitin, eukaryotic initiation factor 5α, and the hypothetical LiHyp1 and LiHyp2 proteins; saponin was used as an adjuvant [155]. The vaccine, composed of 15 μg of ChimeraT plus 15 μg of saponin, was administered in three subcutaneous doses to BALB/c mice, followed by a challenge with L. infantum promastigotes. The vaccine stimulated the production of IFN-γ before and after the infectious challenge and reduced the parasitic load by 80% in the spleen and liver; 90% of parasite reduction was recorded in the bone marrow and lymph node. In the same study, Lage and collaborators used Chimera T, its individual component antigens, and the L. infantum whole cell lysate to stimulate PBMCs from healthy individuals and patients (before and after treatment). It was observed that Chimera T induced higher IFN-γ production in PBMCs compared to the isolated antigens. Interestingly, Chimera T did not stimulate IL-10 production, whereas total L. infantum antigens reduced IFN-γ production and promoted IL-10 secretion. These results suggest that chimeric proteins are promising, as they combine two or more antigens into a single protein that the immune system can recognize, considering the broad and diverse proteome of Leishmania protozoa. Moreover, their ability to stimulate a Th1-type response in human PBMCs highlights their potential use in the prevention of human leishmaniasis.
The antigenic potential of proteins released by Leishmania sp. has been reported [156]. Proteins released by virulent L. infantum promastigotes were divided into two groups: those larger than 75 kDa (LiRic1) and smaller than 37 kDa (LiRic2), which were formulated into two vaccines—5 μg/mL of LiRic1 and LiRic2. BALB/c mice were immunized with three intraperitoneal doses followed by a booster dose seven weeks after the first dose; one week after the last dose, the mice were challenged with L. infantum promastigotes. Immunized animals showed reductions of 50% (LiRic1) and 66% (LiRic2) in splenic parasite burden. In addition, both vaccines stimulated IFN-γ production by CD4+ T cells when stimulated with the LiRic2 antigen. Proteins released by L. shawi promastigotes were separated into three antigenic fractions: >75 kDa (LsPass1), 75–50 kDa (LsPass2), and <50 kDa (LsPass3), which constituted three vaccines—25 μg of LsPass1, LsPass2, or LsPass3, respectively [157]. BALB/c mice were immunized with two intradermal doses and challenged with L. shawi promastigotes [157]. Mice vaccinated with LsPass2 and LsPass3 presented significant reductions in skin parasite burden of 72% and 92%, respectively, compared with non-vaccinated and infected controls. In addition, these mice exhibited a significant reduction in lesion size from the fourth week of infection onward; however, no reduction in parasite burden or lesion size was observed in animals treated with LsPass1 [157]. Although immunization with LsPass1 increased IFN-γ and TNF-α mRNA expression, cytokines considered pro-inflammatory and associated with the activation of antimicrobial responses, this vaccine also stimulated IL-10 mRNA production, which may have contributed to infection persistence and disease progression. Immunization with LsPass2 also induced IFN-γ and TNF-α mRNA expression, accompanied by low IL-10 levels, whereas LsPass3 immunization resulted in moderate IFN-γ and TNF-α production compared to the other formulations. It should be emphasized that a wide variety of proteins are released by Leishmania sp., and that antigenic pools may contain proteins that either stimulate or suppress the host immune response.
Some second-generation vaccines have been licensed for prophylactic use in canine leishmaniasis, including Leishmune®, LeishTec®, CaniLeish®, LetiFend®, and Leish-111f® [158,159,160,161,162]. Leishmune® was produced in Brazil by Zoetis and licensed in 2004; it was composed of the fucose-mannose ligand (FML) of L. donovani plus saponin as an adjuvant [163]. The first report on the efficacy of the use of FML and saponin occurred in 1992 by Palatnik-de-Souza and collaborators in which three intraperitoneal doses were administered to BALB/c mice, each composed of 150 μg of FML and 150 μg of saponin. One week after the last dose, the animals were challenged with L. donovani amastigotes [164]. After 15 days of challenge, it was observed that the animals exhibited 84% less parasites in the liver and a significant increase in the humoral response against the FML antigen compared to unvaccinated animals. This study prompted the subsequent formulation of Leishmune®, as well as the studies related to the efficacy of this vaccine during natural infection. A field study conducted by Nogueira and collaborators observed the vaccine’s protection in dogs, which received three subcutaneous doses [162]. After 11 months of vaccination, 56.7% of non-vaccinated dogs were positive for Leishmania, while 100% of vaccinated dogs showed no clinical signs compatible with leishmaniasis nor detectable parasites in the skin, blood, or lymph nodes. However, in another field study, conducted by Marcondes and collaborators, vaccinated dogs exhibited anti-Leishmania IgG levels similar to the antibody levels of naturally infected dogs. Despite such results, this study was discontinued due to pet owners’ refusal to allow further sample collection, making it impossible to determine the vaccine’s efficacy [165]. Another field study reported that 89% of vaccinated dogs remained asymptomatic and parasite-free after 11 months of vaccination, but 5.1% of vaccinated dogs transmitted the parasite to sandflies during the xenodiagnosis test, contradicting claims that Leishmune® prevents vector transmission [163,166]. The production and commercialization of Leishmune® was discontinued in 2014 due to a lack of evidence of its efficacy in trials.
In the study by Fernandes and collaborators, a field trial was conducted with LeishTec® [166]; a vaccine composed of the recombinant A2 protein from L. donovani amastigotes plus saponin as adjuvant; it was produced in Brazil by Ceva Saúde Animal and licensed in 2007 [163]. LeishTec® was administered subcutaneously in three doses, demonstrating a higher protection rate than Leishmune®, with 92% of vaccinated dogs remaining asymptomatic and undetectable for the parasite; however, 5.4% of vaccinated dogs tested positive for Leishmania transmission in xenodiagnosis [166]. Two additional field trials were conducted during 2016 [167] and 2017 [160]; both using the same three-dose subcutaneous vaccination scheme. In the first study, 63.7% of vaccinated dogs remained asymptomatic and parasite-free [167], while in the second study, the protection rate was 69% [160], but the xenodiagnosis test pointed out that 35.7% of vaccinated animals transmitted the parasite to sandflies [167]. However, similar to Leishmune®, LeishTec® was discontinued by the Ministry of Agriculture and Livestock due to noncompliance with the minimum required antigen content [168].
Letifendi®, produced in Spain by Leti Pharma, contains the recombinant Q protein, which is composed of five antigenic factors derived from four L. infantum proteins [159]. These proteins are widely recognized by the sera of dogs with visceral leishmaniasis [169]. Three antigenic factors originate from ribosomal proteins (LiP0, LiP2a, and LiP2b), while the remaining two are derived from histone 2A (H2A); such proteins contain epitopes that are recognized by B cells, suggesting immunogenicity. The first reports on the protective potential of the recombinant Q vaccine were conducted in mice [170] and dogs [171], prior to the period covered by this review. In these studies, BALB/c mice vaccinated with two doses (15 days apart) of 2 μg of Q protein plus 20 μg of CpG-ODN, and subsequently challenged with L. infantum promastigotes, exhibited 99% reduction in parasite burden in the spleen and liver, along with increased IFN-γ production. In contrast, dogs vaccinated with three intraperitoneal doses (boosters administered 21 and 44 days after the first immunization) containing 4 μg/kg of Q protein plus BCG, and subsequently challenged with L. infantum promastigotes, did not develop clinical signs of disease. Furthermore, only 50% of vaccinated and infected animals showed parasites in lymph nodes at 150 days post-infection, compared with 100% positivity in non-vaccinated, infected dogs. Even before being licensed as a vaccine, a study with the Q protein was conducted using vaccination protocols with different numbers of doses [172]. In this case, it was observed that by changing the dose of Q vaccine to 100 μg and given it as a single dose by subcutaneous route animals presented a higher rate of protection than dogs immunized twice with the same vaccine, since 57% of dogs remained asymptomatic compared to 28% in the group that was immunized with two doses; through histopathological analyses, it was observed that the liver, spleen, and kidneys of the vaccinated dogs exhibited a normal histological morphology. Furthermore, vaccinated animals exhibited a microbicidal response, as evidenced by the elevated production of nitric oxide in lymph node cell cultures compared to non-immunized and infected animals [172]. In another field study, dogs were vaccinated with a single dose of Letifendi® by subcutaneous injection (with a booster dose after one year), and no adverse reactions were observed. These dogs were housed in 19 kennels located in canine leishmaniasis endemic areas of France and Spain. Among the vaccinated dogs, 96.5% remained healthy, with no signs of infection; over the 780-day follow-up period, vaccinated dogs exhibited anti–Q protein IgG2 antibodies, with two significant increases observed after the initial vaccination and the booster dose.
Leish-111f®, also known as MML or LEISH-F1, is a vaccine produced with a chimeric Leishmania antigen, composed of three recombinant antigens: rTSA (L. major), LmSTI1 (L. major), and LeIF (L. braziliensis). The first study that described the efficacy was the MML vaccine to protect mice from the challenge with L. major, which was developed by Coler and collaborators (2002) [173]. It was demonstrated that vaccination of BALB/c mice with three subcutaneous doses containing 10 μg of MML did not induce a humoral immune response. In contrast, the association of MML with the adjuvant MPL-SE induced the production of IgG1 and IgG2a antibodies and stimulated IFN-γ secretion; therefore, this formulation was maintained for the challenge experiment. BALB/c and C57BL/6 mice were immunized following the same protocol and challenged, in the left hind footpad, three weeks after the last dose with metacyclic promastigotes of L. major or L. amazonensis, respectively [173]. Immunization prevented enlargement of the lesion in 80% of the L. major-infected mice and 60% of the L. amazonensis-infected mice. These results demonstrate that the vaccine had a prophylactic effect when formulated with MPL-SE [173]. However, studies combining Leish-111f® with different adjuvants showed that MML + MPL-SE and MML + Adjuprime [90] were ineffective in protecting vaccinated dogs. The animals received three subcutaneous doses at 28-day intervals, and one year after the last dose, the vaccination regimen was repeated. The dogs were kept in open kennels located in an endemic area for human and canine visceral leishmaniasis in Italy. After two years of observation, 37 out of 39 dogs (95%) were infected with L. infantum, and MML failed to induce lymphocyte proliferative responses after the first vaccination schedule. These findings indicate that the antigen and adjuvant doses employed did not confer protection to the animals nor prevent disease progression [90]. It is important to highlight that field studies involve exposure to other pathogens that may increase immunological susceptibility to Leishmania infection, which may explain the discrepancies observed between the two studies, in addition to differences in the animal models used.
Another vaccine for canine leishmaniasis is Canileish®, produced in France by Virbac; this vaccine is also known as LiESP/QA-21, and consists of secreted-excreted proteins from L. infantum and QA-21 as adjuvant [168]. First, this antigen (LiESP) was formulated with adjuvant MDP and tested in dogs [174,175,176]. The animals were immunized with two subcutaneous doses. In two of the three studies, animals were challenged with L. infantum promastigote forms; in both studies, 100% of the vaccinated remained asymptomatic after the challenge, and the parasitic load was undetectable in the bone marrow. The third study was a field trial carried out in regions of southern France where leishmaniasis is highly prevalent [175]. The animals, comprising hunting, guard, and breeding dogs, were maintained in their owners’ households; after two years of follow-up, only 1 out of 165 vaccinated dogs (0.61%) was infected, whereas 12 out of 175 dogs in the placebo group (6.86%) were infected. Moreover, macrophages from vaccinated dogs produced higher levels of NO and IFN-γ compared to those from the placebo group. In a study conducted by Oliva and colleagues, dogs were vaccinated with three doses of the vaccine LiESP/QA-21 and kept in open kennels to allow exposure to sandflies [161]. They observed that the vaccine was well-tolerated by the animals, with the only adverse reaction associated with edema at the injection site. Among the vaccinated animals, 51% were undetectable for Leishmania, and 92% remained free of any disease symptoms.
The second generation of vaccines represents a substantial progress in the development of immunization against leishmaniasis. The isolation and characterization of parasite antigens such as LACK, GP63, histones, and HASPB, and others, provide insights into their immunogenic and antigenic potentials, enabling the selection or even the combination of antigenic factors within a single vaccine formulation. Although the methodology for attaining second-generation has led to the licensing of vaccines for canine leishmaniasis, their efficacy and safety remain unclear, and further studies are required to fill the existing knowledge gaps. Refinement in the selection of antigens and adjuvants, along with a deeper understanding of host-specific immunomodulation, is essential for the development of more effective vaccines against leishmaniasis. Table 5 summarizes the main findings about first-generation vaccines.

5.4. Third Generation Vaccines

Genetic vaccines comprise vaccination strategies performed with deoxyribonucleic acid (DNA vaccines) or messenger ribonucleic acid (mRNA) of the target protein of interest. This technology was initially developed in the 1990s and consisted of intramuscular injections with bacterial plasmid (DNA vaccine) encoding reporter genes, such as chloramphenicol acetyltransferase (CAT), luciferase, and beta-galactosidase, which, after inoculation in transfected cells, were able to express the target protein on the cell surface. In the case of mRNA vaccines, the initial concept was developed by Malone and collaborators, showing that mRNA could also be used to transfect a variety of eukaryotic cells that became able to express a bioactive protein, since these mRNA are given in correct carriers [188].
Basically, the idea of both types of biological products is that a given organism can produce a protein that is missing in biological systems. Although they share a similar technological concept, these vaccines work in different ways. DNA vaccines consist of a bacterial plasmid gene that contains a sequence that will encode an antigen under the control of a eukaryotic promoter, allowing gene expression in transfected cells. In general, plasmids contain an enhancer, a promoter, a transcription termination or polyadenylation signal sequence, and an origin of replication. To facilitate the growth and selection of transfected bacteria, an antibiotic marker is added to the plasmids.
The effectiveness of DNA vaccines depends on different factors, such as the antigen, type of vector, transfection method, number of injections, dose, and adjuvants. Despite several factors controlling the efficacy of a given vaccine, in general, formulations containing DNA vaccines have a similar action mechanism. After being transferred to the host cell (through conventional injection or a gene gun device, for example), the plasmid must enter the nucleus to be transcribed to mRNA. After this step, a bioactive protein will be translated into host cell ribosomes and subjected to post-translational modifications. Depending on the protein features, it can be secreted to the extracellular environment or processed by the major biochemical pathways of histocompatibility, activating the immune system [189].
Therefore, once produced, bioactive protein can be processed by the following pathways: (a) if protein is soluble into the host cell cytoplasm, it will be fragmented by the proteasome, and derived peptides will be presented into the cell surface by major histocompatibility complex I (MHCI), in consequence stimulating CD8+ T lymphocytes, (b) if produced protein is contained in specialized vacuoles of antigen-presenting cells (APC), it will be processed by MHC II machinery and derived peptides will be presented in cell surface in the context of MHC II molecules. In this case, the peptides will trigger CD4+ T cells [190]. Furthermore, DNA-encoded protein can also be processed as an exogenous antigen and presented by (c) MHCI during cross-presentation or (d) be captured by phagocytic cells and presented by MHCII molecules. All these mechanisms can occur together, triggering both cellular and humoral immune responses [191].
In contrast, an mRNA vaccine is not transported to the nucleus, but it is translated into protein directly by ribosomes in the cell cytoplasm. A classical mRNA vaccine contains a 5′ cap, made up of a 7-methylguanosine moiety followed by a triphosphate moiety close to the first nucleotide. This structure protects mRNA vaccines from host enzyme cleavage and regulates splicing. Other structural components of mRNA vaccines are the 5′ and 3′ untranslated regions (UTRs) that are positioned between the gene of interest and both the 5′ and 3′ ends. These regions are not translated into protein but regulate mRNA expression, by having an important role in recognizing mRNA by ribosomes; furthermore, such areas help the post-transcriptional modification of mRNA. Finally, the sequence of interest is added to the RNA vaccine, followed by the poly(A) tail, which is essential to protect RNA from degradation. To protect the RNA vaccine from degradation by innate mechanisms of immunity, vaccines are frequently encapsulated in vesicles containing common phospholipids [192].
Inside a target cell, the mRNA vaccine is going to be translated into proteins by the ribosomes. Depending on the signal peptide in the antigen, the protein will be processed by the MHC I machinery and the derived antigens presented to CD8+ cytotoxic T cells. In APC, the protein is directed to the MHC II compartment, and peptides will be presented on the surface of the APC to CD4+ T cells. Although mRNA vaccines have emerged as a promising immunoprophylactic strategy in the COVID era [102], this strategy was poorly explored in leishmaniasis, and thus, a great focus will be given to DNA vaccines in this section.
In leishmaniasis, the first gene to be assayed as a genetic vaccine was gp63. This gene was inserted into the plasmid pcDNAI, and the DNA vaccine produced was used to immunize BALB/c mice intramuscularly in the skeletal muscle of the thigh with 100 μg of vaccine. This study was a significant landmark for the development of DNA vaccines because it confirmed that immunized animals were able to express the bioactive protein in the cell membrane of muscle cells; in addition, this vaccine was immunogenic, considering the significant stimulation of the Th1 immune response. Importantly, this vaccine partially protected immunized animals and decreased cutaneous parasitism 100 times compared to the control [193].
Other studies also investigated the potential of gp63 in different formulations and vaccination strategies. The protective potential of the DNA vaccine pcDNA3.1-gp63 was compared with the recombinant protein as well as the heterologous prime-boost strategy, constituted with DNA vaccine and recombinant protein plus CpG adjuvant during the challenge performed with L. major in BALB/c mice. In general, it was observed that the heterologous prime-boost strategy, where animals were primed with two intramuscular immunization with pcDNA3.1-gp63 (50 μg/dose, two doses) plus CpG and boosted twice with recombinant gp63 plus CpG, presented the highest protective potential to BALB/c mice, furthermore, animals responded in a Th1 manner, considering the increase in IgG2a over IgG1 isotype levels. In contrast, other immunization strategies that used only pcDNA3.1-gp63 or recombinant protein in the presence of CpG adjuvant also exhibited protective properties; however, only the prime-boost method reduced hepatic and splenic parasitism in a long-term study, suggesting that, in addition to immunogenicity and protection, prime-boost immunization conferred immunological memory to immunized mice. This method potentiates a specific immune response because a DNA vaccine induces high-avidity T cells while recombinant protein boosts the immune system, allowing the proliferation of highly specific and active T cells [194]. The same vaccine also could restrain parasitism in BALB/c mice with visceral leishmaniasis caused by L. donovani; furthermore, this partial protection was associated with elevation in lymphoproliferative and cytotoxic responses, as well as IFN-γ and IL-2 cytokines [195]. Possibly, partial protection observed in such studies may be related to the maintenance of elevated levels of IL-4 and IL-10 [196]. In experiments using a gene gun, it was demonstrated that pcDNA3.1-gp63, given twice, was highly immunogenic and protective to BALB/c mice challenged with L. mexicana compared to the intramuscular route immunization; furthermore, this protection was associated with an increase in CD8+ T cell cytotoxicity [197].
In contrast, the genetic fragment of gp63, comprising the sequence between 138–360aa of this antigen, was cloned into the pVAX1 plasmid and given to BALB/c mice. Studies related to immunogenicity showed that this epitope-based vaccine increased the levels of both IgG1 and IgG2a isotypes, suggesting a mixed immune response. However, after challenge with L. infantum, a potent Th1 immune response was developed, with the participation of CD4+ and CD8+ T lymphocytes, as well as IFN-γ, IL12, and TNF-α. Additionally, a progressive decrease in inflammatory infiltrate and granulomas was observed along with a significant reduction in parasitism [198].
Another antigen administered as a third-generation vaccine is the Leishmania homologue of receptor for activated C kinase (LACK), also called p36. It is a 36-KDa protein highly conserved in the Leishmania genus and is expressed in both stages of the parasite. During the relationship between parasite and host, LACK plays an important role in the establishment of the Th2 immune response, ensuring the persistence of Leishmania in the host cell [199]. Therefore, considering that LACK is conserved in the Leishmania genus and plays a vital role in the establishment of infection, it could be an interesting target to develop a pan-vaccine for leishmaniasis.
In 1997, Gurunathan and colleagues cloned a truncated LACK sequence into the pcDNA3 plasmid and utilized it to immunize BALB/c mice in the hind footpad with 100 μg of vaccine by subcutaneous route with or without a eukaryotic expression vector carrying IL-12. In this case, it was observed that animals challenged with L. major promastigotes exhibited long-lasting protection, considering that the infection was controlled for 20 weeks after challenge. Furthermore, immunized mice controlled the development of lesions at sites distant to the place of immunization, suggesting that systemic protection as well as a significant development of immunological memory, which was associated with the CD8+ T lymphocyte expansion, was prompted after immunization with pcDNA3-LACK vaccine [200,201]. In contrast to this study, it was demonstrated that LACK cloned into the pcDNA3.1 vaccine expression vector does not protect BALB/c mice from the challenge with L. donovani, despite its immunogenic properties in animal models [202]. The same pattern was observed in mice immunized intramuscularly with a DNA vaccine produced with LACK cloned into pCI-neo and challenged with L. chagasi [203].
In experimental American cutaneous leishmaniasis, it was observed that the LACK gene cloned into pcI-neo administered intranasally twice (30 μg/dose), exhibited prophylactic efficacy in BALB/c mice challenged with the L. amazonensis parasite. This genetic vaccine significantly restricted parasite growth and, after 5 months of challenge, elevated levels of IFN-γ in both mucosa and draining lymph nodes, suggesting that this DNA vaccine elicited long-lasting immunity in immunized animals [204]. This same formulation and dose also administered by the mucosal route protected BALB/c mice from challenge with L. infantum, which developed significant Th1 and Th2 immune responses, along with a reduction in IL-10 levels, which is essential to ensure protective immunity in visceral leishmaniasis [205]. Using hamsters, which is an animal model that resembles the natural infection of VL more significantly, it was proven that this experimental vaccine also protected animals challenged with L. infantum [206]. More recently, it was demonstrated that this same vaccine formulated in crosslinked chitosan microparticles was safe after administration to mice intranasally, and protection in models of visceral and cutaneous leishmaniasis was potentized compared to the free vaccine [207,208], suggesting that nanocarriers can be a significant platform for vaccine delivery.
In consideration of the high immunogenicity and protective properties of this vaccine, a heterologous prime-boost strategy was developed, in which the LACK gene was inserted into the pCI-neo plasmid and into an attenuated modified virus Ankara (MVA). LACK-pCI-neo was injected intradermally, and two weeks later, recombinant Ankara virus expressing LACK was injected intraperitoneally. This strategy increased the levels of LACK-specific IFN-g-secreting cells as well as IFN-γ and TNF-α-producing CD4+ and CD8+ T lymphocytes. Despite the Th1 immune response elicited by this strategy, elevated IL-4 levels were detected, suggesting that this vaccine and immunization strategy is highly immunogenic but causes a mixed immune response in the host, which possibly accounted for the partial protection observed after challenge with L. major, however, this protection was maintained during 17 weeks post challenge, suggesting that the vaccine elicited immunological memory [209]. In the hamster model of VL caused by L. infantum, it was demonstrated that the same strategy triggers protection [148].
A similar pattern was observed in canine visceral leishmaniasis; in this case, the beagle dogs were first immunized with 100 μg of the plasmid vector pORT-LACK and 15 days later with 108 pfu of MVA-LACK by the subcutaneous route. This immunization strategy induced a reduction in clinical symptoms as well as hepatic parasitism; however, did not cause a sterile cure in animals [210]. LACK was also inserted into the non-replicative antibiotic-resistant-free pPAL plasmid, and then used to immunize beagle dogs. pPAL-LACK vaccine was immunogenic and induced a significant increase in a Th1 immune response; however, animals challenged with L. infantum exhibited only partial protection after immunization, possibly an imbalance between IFN-γ and IL-10 causes this partial protection [211].
Another well-studied target for the development of genetic vaccines is leishmanial ribosomal proteins. In general, the ribosome translates genetic information into proteins that have different biological roles in parasites. The ribosome structure consists of four ribosomal RNA molecules as well as different associated ribosomal proteins that form large and small ribosomal subunits [212]. In the context of immunization, the development of vaccines containing ribosomal genes or even proteins [213] has been considered a significant strategy, because many of these antigens are conserved in the Leishmania genus [214]; therefore, a multivalent vaccine can be produced.
The first ribosomal protein studied as a genetic vaccine was the acidic ribosomal protein P0 of L. infantum, where the genetic sequence was cloned into pcDNA3 (pcDNA3-LiP0). This genetic vaccine was inoculated intramuscularly in the quadriceps (50 μg per leg) of BALB/c mice twice. Fourteen days after the last dose, the animals were challenged with L. major promastigotes in the footpad. Animals immunized with this genetic vaccine exhibited a delay in the progression of the cutaneous lesion and a reduction in parasitism. Furthermore, the protective mechanism of this vaccine is related to the priming of CD4+ and CD8+ T lymphocytes as well as high production of IFN-γ [215]. This same vaccine was assayed in visceral experimental leishmaniasis caused by L. infantum in golden hamsters; in this regard, a similar protective pattern was observed, where animals that received three doses of pcDNA3-LiP0 intramuscularly (100 μg/dose) exhibited a significant reduction in hepatic and splenic parasitism along with upregulation of a Th1 immune response, furthermore, the liver and spleen presented fewer histopathological changes compared to the infected control [216].
Another gene assayed as a DNA vaccine was the ribosomal P1 gene. The open reading frame of the L. donovani ribosomal P1 gene was cloned into the pVAX1 plasmid (pVAX1-P1). The hamsters were immunized intramuscularly once or twice and challenged with L. donovani promastigotes intracardially two weeks after the last dose. Compared to unvaccinated infected animals, vaccinated animals with pVAX1-P1 were observed to have reduced liver and spleen weight that was correlated with a low parasitism and a high ratio between IFN-γ/IL-4 and IL-12/IL-10, as well as proliferation of splenocytes [72].
More recently, our research group studied the immunogenicity of the S20 ribosomal protein, whose open genetic reading frame was inserted into the pVAX1 vector. This vaccine was administered in BALB/c mice intramuscularly, three times in a 2-week interval. It was observed that BALB/c mice produced antibodies that specifically recognized a protein with 14 kDa, corresponding to the molecular mass of S20 ribosomal protein, suggesting that immunized animals expressed a bioactive protein. Immunologically, increased frequencies of T lymphocytes were detected, which was correlated with a significant enhancement of TNF-α-producing CD8+ T lymphocytes, suggesting that this genetic vaccine has stimulated cytotoxic T lymphocytes. Despite that, it was observed that IFNγ-producing CD4+ and CD8+ T cells decreased by six and seven times in comparison to the control, suggesting that this antigen has immunosuppressive potential (Passero et al., data not published). Furthermore, peritoneal macrophages from the immunized group were more permissive to L. (L.) amazonensis infection than controls, confirming that this antigen has suppressive potential (Figure 7).
Another class of antigens investigated as genetic vaccines is related to the antioxidant machinery of Leishmania sp. This machinery is a set of antioxidant enzymes responsible to detoxify reactive oxygen and nitrogen species, such as hydrogen peroxide, hydroxyl radicals, superoxide anion, nitric oxide, among others, which are capable of damaging lipids, proteins, and nucleic acids, leading parasites to death; in general, these molecules are produced during an immune response or even as a product of drug metabolism [217].
In this regard, it has been well established that the antioxidant machinery of Leishmania, as well as other tripanosomatids, is complex and involves the participation of different enzymes located in several compartments. Iron superoxide dismutase (SOD) detoxifies the superoxide radical into oxygen and hydrogen peroxide, which in turn is detoxified by other enzymes with peroxidase activity, such as tryparedoxin peroxidase (TR). In the context of genetic vaccination, it was demonstrated that TR inserted in the plasmid pcDNA3 (pcDNA3-TR) and in a modified vaccinia virus Ankara (MVA-TR) was immunogenic to BALB/c mice. In this case, the animals were subcutaneously immunized in the rump with 100 μg of pcDNA3-TR at a 3-week interval. Five weeks later, the animals were boosted by the intravenous route with 1 × 106 PFU of MVA-TR. In general, immunization was observed to cause a significant increase in IFN-γ levels that was correlated with partial protection after challenge with L. major. Additionally, it was observed that this strategy induced long-lasting protection, considering that 16 weeks after boost, BALB/c mice still partially controlled lesion development, which was associated with increased IFN-γ and IL-4 production [218].
The same prime-boost strategy using the DNA vaccine and recombinant MVA was employed in the infection caused by L. (V.) panamensis. In this case, TR was cloned into pVAX-1. This strategy showed that this vaccine increased the frequencies of IFN-γ-producing CD4+ and CD8+ T cells, which accounted for a reduced size of the cutaneous lesion in challenge mice as well as low parasite loads in the skin and lymph nodes. Furthermore, this vaccine seems to stimulate CD8+ T lymphocyte clones, considering that their depletion led to a rapid progression of the lesion; in contrast, the depletion of CD4+ T lymphocytes did not alter the protective effect of vaccination [219].
Due to the immunogenicity and protective potential of the TR vaccination, its immunogenicity was evaluated in outbred dogs. In this case, it was demonstrated that the prime-boost strategy using DNA vectors and MVA was safe and immunogenic for dogs, considering that immunized animals produced higher levels of IFN-γ and IgG2a, pointing to a specific and significant Th1 immune response, responsible for activating macrophages [218].
The SOD enzyme is an important molecule responsible for the conversion of superoxide radicals into oxygen and hydrogen peroxide. SOD requires a metal cofactor, such as iron, in the active site to become active [220]. Iron-superoxide dismutase is classified according to the cytoplasmic compartment in which it accumulates. SOD-A and SOD-C can be found in the mitochondria, while SOD-B1 and SOD-B2 can be found in the glycosomes [221,222,223]. This enzyme is a promising antigen to be formulated as a leishmania vaccine because it is absent in humans; thus, severe adverse reactions or even autoimmune reactions are not expected during the vaccination process. Despite work dealing with second-generation vaccines that employ SOD as a prototype vaccine, few works have explored the ability of this class of antigens to be used as a third-generation vaccine.
In this regard, it was demonstrated that a prime-boost strategy performed with a DNA vaccine and recombinant SOD protein was performed in BALB/c mice. Animals were injected intramuscularly with two doses of pcDNA (100 μg plus 25 μg of CpG ODN 1826/animal) containing L. donovani. After 15 days, the animals received one dose of recombinant protein (12.5 μg plus 25 μg of CpG ODN 1826/animal) subcutaneously. It was observed that pcDNA-SOD was immunogenic to mice, inducing a high level of anti-SOD IgG, especially IFN-γ and IgG2a in pcDNA-SOD and pcDNA-GMCSF-SOD groups. Furthermore, enhancement of TNF-α- and IL-2-producing CD4+ T lymphocytes along with T cytotoxic 1 and 2 cell subsets was observed in both groups, culminating in a partial protection after challenge with L. major, as judged by the size of lesions of animals [224]. Although IL-10 levels have been reduced during infection with L. major in immunized animals, the amount still detected should be enough to inhibit the Th1 immune response, allowing parasites to escape from an effective immune response and survive in the host.
Another study showed that the SOD gene inserted in the pVAX1 plasmid (pVAX1-SOD) was also immunogenic when injected three times (100 μg/animal) intramuscularly in BALB/c mice. In this work, animals that received three doses of pVAX1-SOD exhibited high frequencies of IFN-γ-producing CD4+ T cells as well as IFN-γ and IL-4-producing CD8+ T lymphocytes, along with high levels of IgG2a isotype, indicating a protective immune response. After challenge, L. amazonensis parasites abrogated the expansion of IFN-γ-producing CD4+ T cells, observed in immunogenicity studies, but also led to an expansion of IL4-producing CD4+ T lymphocytes. In contrast, a significant expansion of IFN-γ-producing CD8+ T cells was observed, suggesting that partial protection was achieved by the cytotoxic subset of T cells [225]. Another antioxidant enzyme, the thiol-specific antioxidant, formulated as a DNA vaccine, also protected partially challenged mice, possibly due to a significant increase in IL-4 cytokine levels [226,227].
Although antioxidant enzymes seem to be excellent alternatives for producing third-generation vaccines, considering their immunogenicities and the absence of such genes in humans to minimize toxic events, the parasites were able to evade the immune response triggered by experimental immunizations. Possibly, the antioxidant enzyme and other important virulence factors present in viable parasites inhibited the inflammatory environment triggered by just one specific antigen.
From the perspective of DNA vaccines, different studies also investigated the immunogenicity and protective potential of bivalent or multivalent DNA vaccines in preclinical studies using mice, hamsters, or dogs as hosts of cutaneous or visceral species of Leishmania. Examples of such vaccines are TSA and LmSTI1—L. major homolog to the eukaryotic stress-inducible protein [228]; PSA-2, peroxidoxin/TSA, STI1, ARP-1 along with a cocktail of histones [229]; LACK-TSA [227]; KMP11, TSA, CPA, CPB and P74 sequences [230]; and KMPII, TRYP, LACK and GP63 [231]. This strategy is interesting because it is possible to put together different genes or genetic fragments, theoretically assuring protection against different species of Leishmania in hosts with different HLA alleles, therefore, a DNA vaccine containing multiple genes or genic segments would induce a protective immunity in a broad range of individuals [232,233], suggesting that this is a significant strategy to develop a protective vaccine to combat all forms of leishmaniasis. Despite the benefits of producing such DNA vaccines, studies have already demonstrated that only partial protection was achieved after challenge with viable parasites or, in the same cases, failed to protect dogs from an experimental challenge [218]. Table 6 summarizes the main findings on third-generation vaccines.

6. Conclusions

This revision covered different aspects of leishmaniasis, summarizing studies related to the relationship between parasites and hosts, immunology, clinical manifestations, experimental models, adjuvants, and vaccines. By describing these areas of knowledge, it was possible to highlight many elegant works developed toward understanding this complex infectious disease. Despite these advances, in some cases, it is not possible to predict the type of disease that a parasite species will cause during a natural infection in humans. This is, in fact, an association of factors, such as the infecting parasite species (and strain), vector species, the physiological status of the vertebrate host, and genetics. Together, these factors determine the course of the disease and the expression of its clinical form.
At the same time, such elements can also influence the efficacy of vaccines in biological systems. It is also important to mention that Leishmania genus exhibits a great biodiversity of species, which is associated with molecular diversity; this means that phenotypic differences exist between immunogenic proteins, impacting the immunogenicity and protective potential of vaccines. Furthermore, the existence of strains with atypical behavior, such as L. infantum chagasi causing non-ulcerated cutaneous leishmaniasis in Central America, negatively impacts the development of a universal vaccine.
Regarding immunizations, animals are initially injected with a specific antigen and can respond with a significant protective immune response. However, after a challenge with an artificially increased number of parasites, the immune system must struggle with a multitude of parasitic antigens, making it hard to respond specifically, rapidly, and protectively to a single antigen. In this review, all described immunogens conferred partial or no protection. Could these responses be caused by an excessive amount of parasitic antigen in these biological systems? Although these experimental models are important and have contributed to solving immunological puzzles related to leishmaniasis, it would be beneficial to develop new models that better resemble human disease.
Concerning Leishmania vaccines, this review highlights that conserved subunit vaccines are significant, as some important examples of commercialized vaccines originate from the second generation of vaccines. For instance, immunogens employing saponin as an adjuvant are notable at protecting dogs from the infection. In fact, studies on malaria vaccines also indicate that second-generation vaccines can be safely administered to humans and are protective, as evidenced by the development of two important immunogens approved by the WHO to block malaria transmission in humans. Finally, further studies on Leishmania vaccine should align the biology of natural transmission with the principle of a strong and long-lasting cellular immunity, which certainly would block the transmission of Leishmania parasites between invertebrate and vertebrate hosts.

Author Contributions

Conceptualization, L.F.D.P. and M.D.L.; software, I.N.C., G.V.A.F., and S.S.d.L.M.; investigation, L.F.D.P., I.N.C., G.V.A.F., S.S.d.L.M., and M.D.L. resources, L.F.D.P. and M.D.L.; data curation, L.F.D.P. and M.D.L.; writing—original draft preparation, L.F.D.P., I.N.C., G.V.A.F., S.S.d.L.M., and M.D.L.; writing—L.F.D.P., I.N.C., G.V.A.F., S.S.d.L.M., and M.D.L.; visualization, L.F.D.P. and M.D.L.; supervision, L.F.D.P. and M.D.L.; project administration, L.F.D.P. and M.D.L.; funding acquisition, L.F.D.P. and M.D.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by São Paulo State Research Foundation (FAPESP 2023/01641-1) and LIM50-FMUSP. The authors also thank FAPESP for the fellowship awarded to I.N.C., S.S.L.N., and G.V.A.F., and the National Council for Scientific and Technological Development (CNPq) for the grant awarded to L.F.D.P. and M.D.L.

Data Availability Statement

Data are available upon request.

Acknowledgments

The authors acknowledge all researchers around the world working in the field of Leishmania vaccines, which was the basis to produce this work.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. WHO. WHO/Leishmaniasis. Available online: https://www.who.int/news-room/fact-sheets/detail/leishmaniasis (accessed on 30 September 2025).
  2. Passero, L.F.D.; Cruz, L.A.; Santos-Gomes, G.; Rodrigues, E.; Laurenti, M.D.; Lago, J.H.G. Conventional Versus Natural Alternative Treatments for Leishmaniasis: A Review. Curr. Top. Med. Chem. 2018, 18, 1275–1286. [Google Scholar] [CrossRef] [PubMed]
  3. Sacks, D.L.; Brodin, T.N.; Turco, S.J. Developmental Modification of the Lipophosphoglycan from Leishmania Major Promastigotes during Metacyclogenesis. Mol. Biochem. Parasitol. 1990, 42, 225–233. [Google Scholar] [CrossRef] [PubMed]
  4. Conde, L.; Maciel, G.; de Assis, G.M.; Freire-de-Lima, L.; Nico, D.; Vale, A.; Freire-de-Lima, C.G.; Morrot, A. Humoral Response in Leishmaniasis. Front. Cell Infect. Microbiol. 2022, 12, 1063291. [Google Scholar] [CrossRef]
  5. Bogdan, C.; Röllinghoff, M. The Immune Response to Leishmania: Mechanisms of Parasite Control and Evasion. Int. J. Parasitol. 1998, 28, 121–134. [Google Scholar] [CrossRef] [PubMed]
  6. de Menezes, J.P.; Saraiva, E.M.; da Rocha-Azevedo, B. The Site of the Bite: Leishmania Interaction with Macrophages, Neutrophils and the Extracellular Matrix in the Dermis. Parasit. Vectors 2016, 9, 264. [Google Scholar] [CrossRef]
  7. Ritter, U.; Osterloh, A. A New View on Cutaneous Dendritic Cell Subsets in Experimental Leishmaniasis. Med. Microbiol. Immunol. 2007, 196, 51–59. [Google Scholar] [CrossRef]
  8. Carvalho, A.K.; Carvalho, K.; Passero, L.F.D.; Sousa, M.G.T.; da Matta, V.L.R.; Gomes, C.M.C.; Corbett, C.E.P.; Kallas, G.E.; Silveira, F.T.; Laurenti, M.D. Differential Recruitment of Dendritic Cells Subsets to Lymph Nodes Correlates with a Protective or Permissive T-Cell Response during Leishmania (Viannia) Braziliensis or Leishmania (Leishmania) Amazonensis Infection. Mediators Inflamm. 2016, 2016, 1–12. [Google Scholar] [CrossRef]
  9. Ritter, U.; Meißner, A.; Scheidig, C.; Körner, H. CD8α- and Langerin-negative Dendritic Cells, but Not Langerhans Cells, Act as Principal Antigen-presenting Cells in Leishmaniasis. Eur. J. Immunol. 2004, 34, 1542–1550. [Google Scholar] [CrossRef]
  10. Silveira, F.T.; Muller, S.R.; de Souza, A.A.A.; Lainson, R.; Gomes, C.M.C.; Laurenti, M.D.; Corbett, C.E.P. Revisão Sobre a Patogenia Da Leishmaniose Tegumentar Americana Na Amazônia, Com Ênfase à Doença Causada Por Leishmania (V.) Braziliensis e Leishmania (L.) Amazonensis. Rev. Para. De Med. 2008, 22, 9–20. [Google Scholar]
  11. Aranha, F.C.S.; Ribeiro, U.; Basse, P.; Corbett, C.E.P.; Laurenti, M.D. Interleukin-2-Activated Natural Killer Cells May Have a Direct Role in the Control of Leishmania (Leishmania) amazonensis Promastigote and Macrophage Infection. Scand. J. Immunol. 2005, 62, 334–341. [Google Scholar] [CrossRef]
  12. Passero, L.F.D.; Marques, C.; Vale-Gato, I.; Corbett, C.E.P.; Laurenti, M.D.; Santos-Gomes, G. Histopathology, Humoral and Cellular Immune Response in the Murine Model of Leishmania (Viannia) shawi. Parasitol. Int. 2010, 59, 159–165. [Google Scholar] [CrossRef]
  13. Hurdayal, R.; Brombacher, F. The Role of IL-4 and IL-13 in Cutaneous Leishmaniasis. Immunol. Lett. 2014, 161, 179–183. [Google Scholar] [CrossRef]
  14. Passero, L.F.D.; Sacomori, J.V.; Corbett, C.E.P.; Laurenti, M.D. Response of CD4+ and CD8+ T Lymphocytes in the Evolution of Leishmania (Viannia) shawi Infection. Comp. Clin. Pathol. 2012, 21, 521–526. [Google Scholar] [CrossRef]
  15. Brewig, N.; Kissenpfennig, A.; Malissen, B.; Veit, A.; Bickert, T.; Fleischer, B.; Mostböck, S.; Ritter, U. Priming of CD8+ and CD4+ T Cells in Experimental Leishmaniasis Is Initiated by Different Dendritic Cell Subtypes. J. Immunol. 2009, 182, 774–783. [Google Scholar] [CrossRef] [PubMed]
  16. Banerjee, A.; Bhattacharya, P.; Joshi, A.B.; Ismail, N.; Dey, R.; Nakhasi, H.L. Role of Pro-Inflammatory Cytokine IL-17 in Leishmania Pathogenesis and in Protective Immunity by Leishmania Vaccines. Cell Immunol. 2016, 309, 37–41. [Google Scholar] [CrossRef] [PubMed]
  17. Bacellar, O.; Faria, D.; Nascimento, M.; Cardoso, T.M.; Gollob, K.J.; Dutra, W.O.; Scott, P.; Carvalho, E.M. Interleukin 17 Production among Patients with American Cutaneous Leishmaniasis. J. Infect. Dis. 2009, 200, 75–78. [Google Scholar] [CrossRef] [PubMed]
  18. Gonzalez, K.; Calzada, J.E.; Corbett, C.E.P.; Saldaña, A.; Laurenti, M.D. Involvement of the Inflammasome and Th17 Cells in Skin Lesions of Human Cutaneous Leishmaniasis Caused by Leishmania (Viannia) panamensis. Mediators Inflamm. 2020, 2020, 9278931. [Google Scholar] [CrossRef]
  19. Francesquini, F.C.; Silveira, F.T.; Passero, L.F.D.; Tomokane, T.Y.; Carvalho, A.K.; Corbett, C.E.P.; Laurenti, M.D. Salivary Gland Homogenates from Wild-Caught Sand Flies Lutzomyia flaviscutellata and Lutzomyia (Psychodopygus) Complexus Showed Inhibitory Effects on Leishmania (Leishmania) amazonensis and Leishmania (Viannia) braziliensis Infection in BALB/c Mice. Int. J. Exp. Pathol. 2014, 95, 418–426. [Google Scholar] [CrossRef]
  20. de Araújo Albuquerque, L.P.; da Silva, A.M.; de Araújo Batista, F.M.; de Souza Sene, I.; Costa, D.L.; Costa, C.H.N. Influence of Sex Hormones on the Immune Response to Leishmaniasis. Parasite Immunol. 2021, 43, e12874. [Google Scholar] [CrossRef]
  21. Sakthianandeswaren, A.; Foote, S.J.; Handman, E. The Role of Host Genetics in Leishmaniasis. Trends Parasitol. 2009, 25, 383–391. [Google Scholar] [CrossRef]
  22. de Queiroz, N.M.G.P.; da Silveira, R.C.V.; de Noronha, A.C.F.; Oliveira, T.M.F.S.; Machado, R.Z.; Starke-Buzetti, W.A. Detection of Leishmania (L.) Chagasi in Canine Skin. Vet. Parasitol. 2011, 178, 1–8. [Google Scholar] [CrossRef] [PubMed]
  23. Hommel, M.; Jaffe, C.L.; Travi, B.; Milon, G. Experimental Models for Leishmaniasis and for Testing Anti-Leishmanial Vaccines. Ann. Trop. Med. Parasitol. 1995, 89, 55–73. [Google Scholar] [CrossRef] [PubMed]
  24. Alvar, J.; Cañavate, C.; Molina, R.; Moreno, J.; Nieto, J. Canine Leishmaniasis. In Advances in Parasitology; Academic Press: Cambridge, MA, USA, 2004; Volume 57, pp. 1–88. [Google Scholar]
  25. Kumar, R.; Nylén, S. Immunobiology of Visceral Leishmaniasis. Front. Immunol. 2012, 3, 251. [Google Scholar] [CrossRef] [PubMed]
  26. Maia, C.; Nunes, M.; Cristóvão, J.; Campino, L. Experimental Canine Leishmaniasis: Clinical, Parasitological and Serological Follow-Up. Acta Trop. 2010, 116, 193–199. [Google Scholar] [CrossRef]
  27. Laurenti, M.D.; Rossi, C.N.; Matta, V.L.R.d.; Tomokane, T.Y.; Corbett, C.E.P.; Secundino, N.F.C.; Pimenta, P.F.P.; Marcondes, M. Asymptomatic Dogs Are Highly Competent to Transmit Leishmania (Leishmania) infantum Chagasi to the Natural Vector. Vet. Parasitol. 2013, 196, 296–300. [Google Scholar] [CrossRef]
  28. Melby, P.C.; Chandrasekar, B.; Zhao, W.; Coe, J.E.; Teitelbaum, R.; Hariprashad, J.; Murray, H.W. The Hamster as a Model of Human Visceral Leishmaniasis: Progressive Disease and Impaired Generation of Nitric Oxide in the Face of a Prominent Th1-like Cytokine Response. J. Immunol. 2001, 166, 1912–1920. [Google Scholar] [CrossRef]
  29. Miao, J.; Chard, L.S.; Wang, Z.; Wang, Y. Syrian Hamster as an Animal Model for the Study on Infectious Diseases. Front. Immunol. 2019, 10, 2329. [Google Scholar] [CrossRef]
  30. Garg, R.; Dube, A. Animal Models for Vaccine Studies for Visceral Leishmaniasis. Indian J. Med. Res. 2006, 123, 439–454. [Google Scholar]
  31. Corbett, C.E.; Paes, R.A.; Laurenti, M.D.; Andrade Júnior, H.F.; Duarte, M.I. Histopathology of Lymphoid Organs in Experimental Leishmaniasis. Int. J. Exp. Pathol. 1992, 73, 417–433. [Google Scholar]
  32. Laurenti, M.D.; Sotto, M.N.; Corbett, C.E.; da Matta, V.L.; Duarte, M.I. Experimental Visceral Leishmaniasis: Sequential Events of Granuloma Formation at Subcutaneous Inoculation Site. Int. J. Exp. Pathol. 1990, 71, 791–797. [Google Scholar]
  33. Olobo, J.O.; Gicheru, M.M.; Anjili, C.O. The African Green Monkey Model for Cutaneous and Visceral Leishmaniasis. Trends Parasitol. 2001, 17, 588–592. [Google Scholar] [CrossRef] [PubMed]
  34. Porrozzi, R.; Pereira, M.S.; Teva, A.; Volpini, A.C.; Pinto, M.A.; Marchevsky, R.S.; Barbosa, A.A.; Grimaldi, G. Leishmania Infantum-Induced Primary and Challenge Infections in Rhesus Monkeys (Macaca mulatta): A Primate Model for Visceral Leishmaniasis. Trans. R. Soc. Trop. Med. Hyg. 2006, 100, 926–937. [Google Scholar] [CrossRef] [PubMed]
  35. Baneth, G.; Koutinas, A.F.; Solano-Gallego, L.; Bourdeau, P.; Ferrer, L. Canine Leishmaniosis-New Concepts and Insights on an Expanding Zoonosis: Part One. Trends Parasitol. 2008, 24, 324–330. [Google Scholar] [CrossRef] [PubMed]
  36. Awasthi, A.; Mathur, R.K.; Saha, B. Immune Response to Leishmania Infection. Indian. J. Med. Res. 2004, 119, 238–258. [Google Scholar]
  37. Ahmed, S.; Colmenares, M.; Soong, L.; Goldsmith-Pestana, K.; Munstermann, L.; Molina, R.; McMahon-Pratt, D. Intradermal Infection Model for Pathogenesis and Vaccine Studies of Murine Visceral Leishmaniasis. Infect. Immun. 2003, 71, 401–410. [Google Scholar] [CrossRef]
  38. Loría-Cervera, E.N.; Andrade-Narvaez, F.J. Animal models for the study of leishmaniasis immunology. Rev. Inst. Med. Trop. Sao Paulo 2014, 56, 1–11. [Google Scholar] [CrossRef]
  39. Duarte, M.I.; Laurenti, M.D.; Andrade Júnior, H.F.; Corbett, C.E. Comparative Study of the Biological Behaviour in Hamster of Two Isolates of Leishmania Characterized Respectively as L. Major-like and L. Donovani. Rev. Inst. Med. Trop. Sao Paulo 1988, 30, 21–27. [Google Scholar] [CrossRef]
  40. Carneiro, L.A.; Silveira, F.T.; Campos, M.B.; Brígido, M.d.C.d.O.; Gomes, C.M.C.; Corbett, C.E.P.; Laurenti, M.D. Susceptibility of Cebus Apella Monkey (Primates: Cebidae) to Experimental Leishmania (L.) Infantum Chagasi-Infection. Rev. Inst. Med. Trop. Sao Paulo 2011, 53, 45–50. [Google Scholar] [CrossRef]
  41. Costa, S.C.G.; Valle, T.Z. Modelos Experimentais Na Leishmaniose Tegumentar Americana. In Leishmanioses do Continente Americano; Conceição-SILVA, F., Alves, C.R., Eds.; SciELO - Editora FIOCRUZ: Rio de Janeiro, Brazil, 2014; pp. 293–307. ISBN 9788575415689. [Google Scholar]
  42. de Moura, T.R.; Novais, F.O.; Oliveira, F.; Clarêncio, J.; Noronha, A.; Barral, A.; Brodskyn, C.; de Oliveira, C.I. Toward a Novel Experimental Model of Infection To Study American Cutaneous Leishmaniasis Caused by Leishmania Braziliensis. Infect. Immun. 2005, 73, 5827–5834. [Google Scholar] [CrossRef]
  43. Castilho, T.M.; Goldsmith-Pestana, K.; Lozano, C.; Valderrama, L.; Saravia, N.G.; McMahon-Pratt, D. Murine Model of Chronic L. (Viannia) Panamensis Infection: Role of IL-13 in Disease. Eur. J. Immunol. 2010, 40, 2816–2829. [Google Scholar] [CrossRef]
  44. Silva, V.M.G.; Larangeira, D.F.; Oliveira, P.R.S.; Sampaio, R.B.; Suzart, P.; Nihei, J.S.; Teixeira, M.C.A.; Mengel, J.O.; dos-Santos, W.L.C.; Pontes-de-Carvalho, L. Enhancement of Experimental Cutaneous Leishmaniasis by Leishmania Molecules Is Dependent on Interleukin-4, Serine Protease/Esterase Activity, and Parasite and Host Genetic Backgrounds. Infect. Immun. 2011, 79, 1236–1243. [Google Scholar] [CrossRef]
  45. Silveira, F.T.; Lainson, R.; Corbett, C.E. Clinical and Immunopathological Spectrum of American Cutaneous Leishmaniasis with Special Reference to the Disease in Amazonian Brazil: A Review. Mem. Inst. Oswaldo Cruz 2004, 99, 239–251. [Google Scholar] [CrossRef] [PubMed]
  46. Calabrese, K.d.S.; da Costa, S.C.G. Enhancement of Leishmania amazonensis Infection in BCG Non-Responder Mice by BCG-Antigen Specific Vaccine. Mem. Inst. Oswaldo Cruz 1992, 87, 49–56. [Google Scholar] [CrossRef] [PubMed]
  47. de Oliveira Cardoso, F.; de Souza, C.d.S.F.; Mendes, V.G.; Abreu-Silva, A.L.; Gonçalves da Costa, S.C.; Calabrese, K.d.S. Immunopathological Studies of Leishmania amazonensis Infection in Resistant and in Susceptible Mice. J. Infect. Dis. 2010, 201, 1933–1940. [Google Scholar] [CrossRef] [PubMed]
  48. Ji, J.; Sun, J.; Soong, L. Impaired Expression of Inflammatory Cytokines and Chemokines at Early Stages of Infection with Leishmania amazonensis. Infect. Immun. 2003, 71, 4278–4288. [Google Scholar] [CrossRef]
  49. Amaral, V.F.; Ransatto, V.A.O.; Conceição-Silva, F.; Molinaro, E.; Ferreira, V.; Coutinho, S.G.; McMahon-Pratt, D.; Grimaldi, G., Jr. Leishmania amazonensis:The Asian Rhesus Macaques (Macaca Mulatta) as an Experimental Model for Study of Cutaneous Leishmaniasis. Exp. Parasitol. 1996, 82, 34–44. [Google Scholar] [CrossRef]
  50. Laurenti, M.D.; Passero, L.F.D.; Tomokane, T.Y.; Francesquini, F.d.C.; Rocha, M.C.; Gomes, C.M.d.C.; Corbett, C.E.P.; Silveira, F.T. Dynamic of the Cellular Immune Response at the Dermal Site of Leishmania (L.) Amazonensis and Leishmania (V.) Braziliensis Infection in Sapajus Apella Primate. Biomed. Res. Int. 2014, 2014, 134236. [Google Scholar] [CrossRef]
  51. DeKrey, G.K.; Lima, H.C.; Titus, R.G. Analysis of the Immune Responses of Mice to Infection with Leishmania Braziliensis. Infect. Immun. 1998, 66, 827–829. [Google Scholar] [CrossRef]
  52. Boroumand, H.; Badie, F.; Mazaheri, S.; Seyedi, Z.S.; Nahand, J.S.; Nejati, M.; Baghi, H.B.; Abbasi-Kolli, M.; Badehnoosh, B.; Ghandali, M.; et al. Chitosan-Based Nanoparticles Against Viral Infections. Front. Cell Infect. Microbiol. 2021, 11, 643953. [Google Scholar] [CrossRef]
  53. Ratnapriya, S.; Keerti; Sahasrabuddhe, A.A.; Dube, A. Visceral Leishmaniasis: An Overview of Vaccine Adjuvants and Their Applications. Vaccine 2019, 37, 3505–3519. [Google Scholar] [CrossRef]
  54. Dmour, I.; Islam, N. Recent Advances on Chitosan as an Adjuvant for Vaccine Delivery. Int. J. Biol. Macromol. 2022, 200, 498–519. [Google Scholar] [CrossRef]
  55. Di Pasquale, A.; Preiss, S.; Da Silva, F.T.; Garçon, N. Vaccine Adjuvants: From 1920 to 2015 and Beyond. Vaccines 2015, 3, 320–343. [Google Scholar] [CrossRef]
  56. Facciolà, A.; Visalli, G.; Laganà, A.; Di Pietro, A. An Overview of Vaccine Adjuvants: Current Evidence and Future Perspectives. Vaccines 2022, 10, 819. [Google Scholar] [CrossRef] [PubMed]
  57. Iwasaki, A.; Omer, S.B. Why and How Vaccines Work. Cell 2020, 183, 290–295. [Google Scholar] [CrossRef] [PubMed]
  58. Bhowmick, S.; Ravindran, R.; Ali, N. IL-4 Contributes to Failure, and Colludes with IL-10 to Exacerbate Leishmania Donovani Infection Following Administration of a Subcutaneous Leishmanial Antigen Vaccine. BMC Microbiol. 2014, 14. [Google Scholar] [CrossRef] [PubMed]
  59. Wheat, W.H.; Arthun, E.N.; Spencer, J.S.; Regan, D.P.; Titus, R.G.; Dow, S.W. Immunization against Full-Length Protein and Peptides from the Lutzomyia Longipalpis Sand Fly Salivary Component Maxadilan Protects against Leishmania Major Infection in a Murine Model. Vaccine 2017, 35, 6611–6619. [Google Scholar] [CrossRef]
  60. Akbari, M.; Oryan, A.; Hatam, G. Immunotherapy in Treatment of Leishmaniasis. Immunol. Lett. 2021, 233, 80–86. [Google Scholar] [CrossRef]
  61. Shibaki, A.; Katz, S.I. Induction of Skewed Th1/Th2 T-Cell Differentiation via Subcutaneous Immunization with Freund’s Adjuvant. Exp. Dermatol 2002, 11, 126–134. [Google Scholar] [CrossRef]
  62. Billiau, A.; Matthys, P. Modes of Action of Freund’s Adjuvants in Experimental Models of Autoimmune Diseases. J. Leukoc. Biol. 2001, 70, 849–860. [Google Scholar] [CrossRef]
  63. Cerwenka, A.; Lanier, L.L. Ligands for Natural Killer Cell Receptors: Redundancy or Specificity. Immunol. Rev. 2001, 181, 158–169. [Google Scholar] [CrossRef]
  64. McKee, A.S.; Marrack, P. Old and New Adjuvants. Curr. Opin. Immunol. 2017, 47, 44–51. [Google Scholar] [CrossRef]
  65. Yip, H.C.; Karulin, A.Y.; Tary-Lehmann, M.; Hesse, M.D.; Radeke, H.; Heeger, P.S.; Trezza, R.P.; Heinzel, F.P.; Forsthuber, T.; Lehmann, P.V. Adjuvant-Guided Type-1 and Type-2 Immunity: Infectious/Noninfectious Dichotomy Defines the Class of Response. J. Immunol. 1999, 162, 3942–3949. [Google Scholar] [CrossRef] [PubMed]
  66. Jackson, L.R.; Fox, J.G. Institutional Policies and Guidelines on Adjuvants and Antibody Production. ILAR J. 1995, 37, 141–152. [Google Scholar] [CrossRef] [PubMed]
  67. Fan, J.; Jin, S.; Gilmartin, L.; Toth, I.; Hussein, W.M.; Stephenson, R.J. Advances in Infectious Disease Vaccine Adjuvants. Vaccines 2022, 10, 1120. [Google Scholar] [CrossRef] [PubMed]
  68. Luchner, M.; Reinke, S.; Milicic, A. Tlr Agonists as Vaccine Adjuvants Targeting Cancer and Infectious Diseases. Pharmaceutics 2021, 13, 142. [Google Scholar] [CrossRef]
  69. Khabazzadeh Tehrani, N.; Mahdavi, M.; Maleki, F.; Zarrati, S.; Tabatabaie, F. The Role of Montanide ISA 70 as an Adjuvant in Immune Responses against Leishmania major Induced by Thiol-Specific Antioxidant-Based Protein Vaccine. J. Parasit. Dis. 2016, 40, 760–767. [Google Scholar] [CrossRef]
  70. Das, P.; Paik, D.; Naskar, K.; Chakraborti, T. Leishmania Donovani Serine Protease Encapsulated in Liposome Elicits Protective Immunity in Experimental Visceral Leishmaniasis. Microbes Infect. 2018, 20, 37–47. [Google Scholar] [CrossRef]
  71. Allahverdiyev, A.M.; Cakir Koc, R.; Bagirova, M.; Elcicek, S.; Baydar, S.Y.; Oztel, O.N.; Abamor, E.S.; Ates, S.C.; Topuzogullari, M.; Isoglu Dincer, S.; et al. A New Approach for Development of Vaccine against Visceral Leishmaniasis: Lipophosphoglycan and Polyacrylic Acid Conjugates. Asian Pac. J. Trop. Med. 2017, 10, 877–886. [Google Scholar] [CrossRef]
  72. Masih, S.; Arora, S.K.; Vasishta, R.K. Efficacy of Leishmania Donovani Ribosomal P1 Gene as DNA Vaccine in Experimental Visceral Leishmaniasis. Exp. Parasitol. 2011, 129, 55–64. [Google Scholar] [CrossRef]
  73. Petroff, S.A.; Branch, A. Bacillus Calmette-Guérin (B.C.G.): Animal Experimentation and Prophylactic Immunization of Children. Am. J. Public. Health Nations Health 1928, 18, 843–864. [Google Scholar] [CrossRef]
  74. Yamazaki-Nakashimada, M.A.; Unzueta, A.; Berenise Gámez-González, L.; González-Saldaña, N.; Sorensen, R.U. BCG: A Vaccine with Multiple Faces. Hum. Vaccin. Immunother. 2020, 16, 1841–1850. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, J.; Gao, L.; Wu, X.; Fan, Y.; Liu, M.; Peng, L.; Song, J.; Li, B.; Liu, A.; Bao, F. BCG-Induced Trained Immunity: History, Mechanisms and Potential Applications. J. Transl. Med. 2023, 21, 106. [Google Scholar] [CrossRef] [PubMed]
  76. Balodi, D.C.; Anand, A.; Ramalingam, K.; Yadav, S.; Goyal, N. Dipeptidylcarboxypeptidase of Leishmania Donovani: A Potential Vaccine Molecule against Experimental Visceral Leishmaniasis. Cell Immunol. 2022, 375. [Google Scholar] [CrossRef] [PubMed]
  77. Ferreira, J.H.L.; Gentil, L.G.; Dias, S.S.; Fedeli, C.E.C.; Katz, S.; Barbiéri, C.L. Immunization with the Cysteine Proteinase Ldccys1 Gene from Leishmania (Leishmania) Chagasi and the Recombinant Ldccys1 Protein Elicits Protective Immune Responses in a Murine Model of Visceral Leishmaniasis. Vaccine 2008, 26, 677–685. [Google Scholar] [CrossRef]
  78. Nahrevanian, H.; Jafary, S.P.; Nemati, S.; Farahmand, M.; Omidinia, E. Evaluation of Anti-Leishmanial Effects of Killed Leishmania Vaccine with BCG Adjuvant in BALB/c Mice Infected with Leishmania Major MRHO/IR/75/ER. FoliA Parasitol. 2013, 60, 1–6. [Google Scholar] [CrossRef]
  79. Michel, M.Y.; Fathy, F.M.; Hegazy, E.H.; Hussein, E.D.; Eissa, M.M.; Said, D.E. The Adjuvant Effects of IL-12 and BCG on Autoclaved Leishmania Major Vaccine in Experimental Cutaneous Leishmaniasis. J. Egypt Soc. Parasitol. 2006, 36. [Google Scholar]
  80. Ratnapriya, S.; Keerti; Yadav, N.K.; Dube, A.; Sahasrabuddhe, A.A. A Chimera of Th1 Stimulatory Proteins of Leishmania Donovani Offers Moderate Immunotherapeutic Efficacy with a Th1-Inclined Immune Response against Visceral Leishmaniasis. Biomed. Res. Int. 2021, 2021, 8845826. [Google Scholar] [CrossRef]
  81. Shetab Boushehri, M.A.; Lamprecht, A. TLR4-Based Immunotherapeutics in Cancer: A Review of the Achievements and Shortcomings. Mol. Pharm. 2018, 15, 4777–4800. [Google Scholar] [CrossRef]
  82. Schülke, S.; Flaczyk, A.; Vogel, L.; Gaudenzio, N.; Angers, I.; Löschner, B.; Wolfheimer, S.; Spreitzer, I.; Qureshi, S.; Tsai, M.; et al. MPLA Shows Attenuated Pro-Inflammatory Properties and Diminished Capacity to Activate Mast Cells in Comparison with LPS. Allergy 2015, 70, 1259–1268. [Google Scholar] [CrossRef]
  83. Raman, V.S.; Duthie, M.S.; Fox, C.B.; Matlashewski, G.; Reed, S.G. Adjuvants for Leishmania Vaccines: From Models to Clinical Application. Front. Immunol. 2012, 3, 23564. [Google Scholar] [CrossRef]
  84. D’Onofrio, V.; Santana, A.C.; Pauwels, M.; Waerlop, G.; Willems, A.; De Boever, F.; Müller, M.; Sehr, P.; Waterboer, T.; Leroux-Roels, I.; et al. AS04 Drives Superior Cross-Protective Antibody Response by Increased NOTCH Signaling of Dendritic Cells and Proliferation of Memory B Cells. Front. Immunol. 2025, 16, 1623405. [Google Scholar] [CrossRef] [PubMed]
  85. Beran, J. Safety and Immunogenicity of a New Hepatitis B Vaccine for the Protection of Patients with Renal Insufficiency Including Pre-Haemodialysis and Haemodialysis Patients. Expert. Opin. Biol. Ther. 2008, 8, 235–247. [Google Scholar] [CrossRef] [PubMed]
  86. Thakur, A.; Kaur, H.; Kaur, S. Evaluation of the Immunogenicity and Protective Efficacy of Killed Leishmania Donovani Antigen along with Different Adjuvants against Experimental Visceral Leishmaniasis. Med. Microbiol. Immunol. 2015, 204, 539–550. [Google Scholar] [CrossRef] [PubMed]
  87. Bhardwaj, S.; Vasishta, R.K.; Arora, S.K. Vaccination with a Novel Recombinant Leishmania Antigen plus MPL Provides Partial Protection against L. Donovani Challenge in Experimental Model of Visceral Leishmaniasis. Exp. Parasitol. 2009, 121, 29–37. [Google Scholar] [CrossRef]
  88. Skeiky, Y.A.W.; Coler, R.N.; Brannon, M.; Stromberg, E.; Greeson, K.; Thomas Crane, R.; Campos-Neto, A.; Reed, S.G. Protective Efficacy of a Tandemly Linked, Multi-Subunit Recombinant Leishmanial Vaccine (Leish-111f) Formulated in MPL® Adjuvant. Vaccine 2002, 20, 3292–3303. [Google Scholar] [CrossRef]
  89. Fujiwara, R.T.; Vale, A.M.; França Da Silva, J.C.; Da Costa, R.T.; Quetz, J.D.S.; Martins Filho, O.A.; Reis, A.B.; Corrêa Oliveira, R.; Machado-Coelho, G.L.; Bueno, L.L.; et al. Immunogenicity in Dogs of Three Recombinant Antigens (TSA, LeIF and LmSTI1) Potential Vaccine Candidates for Canine Visceral Leishmaniasis. Vet. Res. 2005, 36, 827–838. [Google Scholar] [CrossRef]
  90. Gradoni, L.; Foglia Manzillo, V.; Pagano, A.; Piantedosi, D.; De Luna, R.; Gramiccia, M.; Scalone, A.; Di Muccio, T.; Oliva, G. Failure of a Multi-Subunit Recombinant Leishmanial Vaccine (MML) to Protect Dogs from Leishmania Infantum Infection and to Prevent Disease Progression in Infected Animals. Vaccine 2005, 23, 5245–5251. [Google Scholar] [CrossRef]
  91. Llanos-Cuentas, A.; Calderón, W.; Cruz, M.; Ashman, J.A.; Alves, F.P.; Coler, R.N.; Bogatzki, L.Y.; Bertholet, S.; Laughlin, E.M.; Kahn, S.J.; et al. A Clinical Trial to Evaluate the Safety and Immunogenicity of the LEISH-F1+MPL-SE Vaccine When Used in Combination with Sodium Stibogluconate for the Treatment of Mucosal Leishmaniasis. Vaccine 2010, 28, 7427–7435. [Google Scholar] [CrossRef]
  92. Chen, W.; Jiang, M.; Yu, W.; Xu, Z.; Liu, X.; Jia, Q.; Guan, X.; Zhang, W. CpG-Based Nanovaccines for Cancer Immunotherapy. Int. J. Nanomedicine 2021, 16, 5281–5299. [Google Scholar] [CrossRef]
  93. Bode, C.; Zhao, G.; Steinhagen, F.; Kinjo, T.; Klinman, D.M. CpG DNA as a Vaccine Adjuvant. Expert. Rev. Vaccines 2011, 10, 499–511. [Google Scholar] [CrossRef]
  94. Shi, S.; Zhu, H.; Xia, X.; Liang, Z.; Ma, X.; Vaccine, B.S. Vaccine Adjuvants: Understanding the Structure and Mechanism of Adjuvanticity. Vaccine 2019, 37, 3167–3178. [Google Scholar] [CrossRef] [PubMed]
  95. Vollmer, J.; Krieg, A.M. Immunotherapeutic Applications of CpG Oligodeoxynucleotide TLR9 Agonists. Adv. Drug Deliv. Rev. 2009, 61, 195–204. [Google Scholar] [CrossRef] [PubMed]
  96. Daifalla, N.S.; Bayih, A.G.; Gedamu, L. Leishmania Donovani Recombinant Iron Superoxide Dismutase B1 Protein in the Presence of TLR-Based Adjuvants Induces Partial Protection of BALB/c Mice against Leishmania Major Infection. Exp. Parasitol. 2012, 131, 317–324. [Google Scholar] [CrossRef] [PubMed]
  97. Tafaghodi, M.; Eskandari, M.; Khamesipour, A.; Jaafari, M.R. Alginate Microspheres Encapsulated with Autoclaved Leishmania Major (ALM) and CpG-ODN Induced Partial Protection and Enhanced Immune Response against Murine Model of Leishmaniasis. Exp. Parasitol. 2011, 129, 107–114. [Google Scholar] [CrossRef]
  98. Katzenellenbogen, I. Vaccination against Oriental Sore. Arch. Derm. Syphilol. 1944, 50, 239. [Google Scholar] [CrossRef]
  99. Daifalla, N.S.; Bayih, A.G.; Gedamu, L. Immunogenicity of Leishmania Donovani Iron Superoxide Dismutase B1 and Peroxidoxin 4 in BALB/c Mice: The Contribution of Toll-like Receptor Agonists as Adjuvant. Exp. Parasitol. 2011, 129, 292–298. [Google Scholar] [CrossRef]
  100. Iwasaki, A.; Medzhitov, R. Toll-like Receptor Control of the Adaptive Immune Responses. Nat. Immunol. 2004, 5, 987–995. [Google Scholar] [CrossRef]
  101. Mutwiri, G. TLR9 Agonists: Immune Mechanisms and Therapeutic Potential in Domestic Animals. Vet. Immunol. Immunopathol. 2012, 148, 85–89. [Google Scholar] [CrossRef]
  102. Shafaati, M.; Saidijam, M.; Soleimani, M.; Hazrati, F.; Mirzaei, R.; Amirheidari, B.; Tanzadehpanah, H.; Karampoor, S.; Kazemi, S.; Yavari, B.; et al. A Brief Review on Dna Vaccines in the Era of COVID-19. Future Virol. 2022, 17, 49–66. [Google Scholar] [CrossRef]
  103. Tsoumani, M.E.; Voyiatzaki, C.; Efstathiou, A. Malaria Vaccines: From the Past towards the mRNA Vaccine Era. Vaccines 2023, 11, 1452. [Google Scholar] [CrossRef]
  104. Saini, I.; Joshi, J.; Kaur, S. Leishmania Vaccine Development: A Comprehensive Review. Cell Immunol. 2024, 399–400, 104826. [Google Scholar] [CrossRef]
  105. Khamesipour, A.; Dowlati, Y.; Asilian, A.; Hashemi-Fesharki, R.; Javadi, A.; Noazin, S.; Modabber, F. Leishmanization: Use of an old method for evaluation of candidate vaccines against leishmaniasis. Vaccine 2005, 23, 3642–3648. [Google Scholar] [CrossRef]
  106. Senekji, H.A.; Beattie, C.P. Artificial Infection and Immunization of Man with Cultures of Leishmania Tropica. Trans. R. Soc. Trop. Med. Hyg. 1941, 34, 415–419. [Google Scholar] [CrossRef]
  107. Naggan, L.; Gunders, A.E.; Michaeli, D. Follow-up Study of a Vaccination Programme against Cutaneous Leishmaniasis. Trans. R. Soc. Trop. Med. Hyg. 1972, 66, 239–243. [Google Scholar] [CrossRef] [PubMed]
  108. Nadirn, A.; Javadian, E.; Mohebali, M. The Experience of Leishmanization in the Islamic Republic of Iran. Eastern IRevelent 1997, 3, 284. [Google Scholar]
  109. Ismail, N.; Karmakar, S.; Bhattacharya, P.; Dey, R.; Nakhasi, H.L. Immunization with Leishmania major Centrin Knock-out (LmCen −/−) Parasites Induces Skin Resident Memory T Cells That Plays a Role in Protection against Wild Type Infection (LmWT). J. Immunol. 2019, 202, 196.29. [Google Scholar] [CrossRef]
  110. Laabs, E.M.; Wu, W.; Mendez, S. Vaccination with Live Leishmania major and CpG DNA Promotes Interleukin-2 Production by Dermal Dendritic Cells and NK Cell Activation. Clin. Vaccine Immunol. 2009, 16, 1601–1606. [Google Scholar] [CrossRef]
  111. Mendez, S.; Tabbara, K.; Belkaid, Y.; Bertholet, S.; Verthelyi, D.; Klinman, D.; Seder, R.A.; Sacks, D.L. Coinjection with CpG-Containing Immunostimulatory Oligodeoxynucleotides Reduces the Pathogenicity of a Live Vaccine against Cutaneous Leishmaniasis but Maintains Its Potency and Durability. Infect. Immun. 2003, 71, 5121–5129. [Google Scholar] [CrossRef]
  112. Mizbani, A.; Taheri, T.; Zahedifard, F.; Taslimi, Y.; Azizi, H.; Azadmanesh, K.; Papadopoulou, B.; Rafati, S. Recombinant Leishmania Tarentolae Expressing the A2 Virulence Gene as a Novel Candidate Vaccine against Visceral Leishmaniasis. Vaccine 2009, 28, 53–62. [Google Scholar] [CrossRef]
  113. Breton, M.; Tremblay, M.J.; Ouellette, M.; Papadopoulou, B. Live Nonpathogenic Parasitic Vector as a Candidate Vaccine against Visceral Leishmaniasis. Infect. Immun. 2005, 73, 6372–6382. [Google Scholar] [CrossRef]
  114. Keshavarzian, N.; Noroozbeygi, M.; Haji Molla Hoseini, M.; Yeganeh, F. Evaluation of Leishmanization Using Iranian Lizard Leishmania Mixed With CpG-ODN as a Candidate Vaccine Against Experimental Murine Leishmaniasis. Front. Immunol. 2020, 11. [Google Scholar] [CrossRef] [PubMed]
  115. Daneshvar, H.; Namazi, M.J.; Kamiabi, H.; Burchmore, R.; Cleaveland, S.; Phillips, S. Gentamicin-Attenuated Leishmania Infantum Vaccine: Protection of Dogs against Canine Visceral Leishmaniosis in Endemic Area of Southeast of Iran. PLoS Negl. Trop. Dis. 2014, 8, e2757. [Google Scholar] [CrossRef] [PubMed]
  116. Davoudi, N.; Khamesipour, A.; Mahboudi, F.; McMaster, W.R. A Dual Drug Sensitive L. Major Induces Protection without Lesion in C57BL/6 Mice. PLoS Negl. Trop. Dis. 2014, 8, e2785. [Google Scholar] [CrossRef] [PubMed]
  117. Carrión, J.; Folgueira, C.; Soto, M.; Fresno, M.; Requena, J.M. Leishmania Infantum HSP70-II Null Mutant as Candidate Vaccine against Leishmaniasis: A Preliminary Evaluation. Parasit. Vectors 2011, 4, 150. [Google Scholar] [CrossRef]
  118. Karmakar, S.; Ismail, N.; Oliveira, F.; Oristian, J.; Zhang, W.W.; Kaviraj, S.; Singh, K.P.; Mondal, A.; Das, S.; Pandey, K.; et al. Preclinical Validation of a Live Attenuated Dermotropic Leishmania Vaccine against Vector Transmitted Fatal Visceral Leishmaniasis. Commun. Biol. 2021, 4, 929. [Google Scholar] [CrossRef]
  119. Ismail, N.; Karmakar, S.; Bhattacharya, P.; Sepahpour, T.; Takeda, K.; Hamano, S.; Matlashewski, G.; Satoskar, A.R.; Gannavaram, S.; Dey, R.; et al. Leishmania Major Centrin Gene-Deleted Parasites Generate Skin Resident Memory T-Cell Immune Response Analogous to Leishmanization. Front. Immunol. 2022, 13. [Google Scholar] [CrossRef]
  120. McCall, L.-I.; Zhang, W.-W.; Ranasinghe, S.; Matlashewski, G. Leishmanization Revisited: Immunization with a Naturally Attenuated Cutaneous Leishmania Donovani Isolate from Sri Lanka Protects against Visceral Leishmaniasis. Vaccine 2013, 31, 1420–1425. [Google Scholar] [CrossRef]
  121. Davoudi, N.; Tate, C.A.; Warburton, C.; Murray, A.; Mahboudi, F.; McMaster, W.R. Development of a Recombinant Leishmania Major Strain Sensitive to Ganciclovir and 5-Fluorocytosine for Use as a Live Vaccine Challenge in Clinical Trials. Vaccine 2005, 23, 1170–1177. [Google Scholar] [CrossRef]
  122. Peters, N.C.; Kimblin, N.; Secundino, N.; Kamhawi, S.; Lawyer, P.; Sacks, D.L. Vector Transmission of Leishmania Abrogates Vaccine-Induced Protective Immunity. PLoS Pathog. 2009, 5, e1000484. [Google Scholar] [CrossRef]
  123. Mayrink, W.; Tavares, C.A.P.; de Deus, R.B.; Pinheiro, M.B.; Guimarães, T.M.P.D.; de Andrade, H.M.; da Costa, C.A.; de Toledo, V.d.P.C.P. Comparative Evaluation of Phenol and Thimerosal as Preservatives for a Candidate Vaccine against American Cutaneous Leishmaniasis. Mem. Inst. Oswaldo Cruz 2010, 105, 86–91. [Google Scholar] [CrossRef]
  124. Mutiso, J.M.; Macharia, J.C.; Mutisya, R.M.; Taracha, E. Subcutaneous Immunization against Leishmania Major—Infection in Mice: Efficacy of Formalin-Killed Promastigotes Combined with Adjuvants. Rev. Inst. Med. Trop. Sao Paulo 2010, 52, 95–100. [Google Scholar] [CrossRef] [PubMed]
  125. Germanó, M.J.; Lozano, E.S.; Sanchez, M.V.; Bruna, F.A.; García-Bustos, M.F.; Lochedino, A.L.S.; Salomón, M.C.; Fernandes, A.P.; Mackern-Oberti, J.P.; Cargnelutti, D.E. Evaluation of Different Total Leishmania amazonensis Antigens for the Development of a First-Generation Vaccine Formulated with a Toll-like Receptor-3 Agonist to Prevent Cutaneous Leishmaniasis. Mem. Inst. Oswaldo Cruz 2020, 115, e200067. [Google Scholar] [CrossRef]
  126. Noazin, S.; Modabber, F.; Khamesipour, A.; Smith, P.G.; Moulton, L.H.; Nasseri, K.; Sharifi, I.; Khalil, E.A.G.; Bernal, I.D.V.; Antunes, C.M.F.; et al. First Generation Leishmaniasis Vaccines: A Review of Field Efficacy Trials. Vaccine 2008, 26, 6759–6767. [Google Scholar] [CrossRef] [PubMed]
  127. Handman, E. Leishmaniasis: Current Status of Vaccine Development. Clin. Microbiol. Rev. 2001, 14, 229–243. [Google Scholar] [CrossRef] [PubMed]
  128. Grenfell, R.F.Q.; Marques-da-Silva, E.A.; Souza-Testasicca, M.C.; Coelho, E.A.F.; Fernandes, A.P.; Afonso, L.C.C.; Rezende, S.A. Antigenic Extracts of Leishmania Braziliensis and Leishmania amazonensis Associated with Saponin Partially Protects BALB/c Mice against Leishmania Chagasi Infection by Suppressing IL-10 and IL-4 Production. Mem. Inst. Oswaldo Cruz 2010, 105, 818–822. [Google Scholar] [CrossRef]
  129. Giunchetti, R.C.; Corrêa-Oliveira, R.; Martins-Filho, O.A.; Teixeira-Carvalho, A.; Roatt, B.M.; de Oliveira Aguiar-Soares, R.D.; de Souza, J.V.; das Dores Moreira, N.; Malaquias, L.C.C.; Mota e Castro, L.L.; et al. Immunogenicity of a Killed Leishmania Vaccine with Saponin Adjuvant in Dogs. Vaccine 2007, 25, 7674–7686. [Google Scholar] [CrossRef]
  130. Giunchetti, R.C.; Corrêa-Oliveira, R.; Martins-Filho, O.A.; Teixeira-Carvalho, A.; Roatt, B.M.; Aguiar-Soares, R.D.d.O.; Coura-Vital, W.; de Abreu, R.T.; Malaquias, L.C.C.; Gontijo, N.F.; et al. A Killed Leishmania Vaccine with Sand Fly Saliva Extract and Saponin Adjuvant Displays Immunogenicity in Dogs. Vaccine 2008, 26, 623–638. [Google Scholar] [CrossRef]
  131. Giunchetti, R.C.; Reis, A.B.; da Silveira-Lemos, D.; Martins-Filho, O.A.; Corrêa-Oliveira, R.; Bethony, J.; Vale, A.M.; da Silva Quetz, J.; Bueno, L.L.; França-Silva, J.C.; et al. Antigenicity of a Whole Parasite Vaccine as Promising Candidate against Canine Leishmaniasis. Res. Vet. Sci. 2008, 85, 106–112. [Google Scholar] [CrossRef]
  132. Vélez, I.D.; Gilchrist, K.; Arbelaez, M.P.; Rojas, C.A.; Puerta, J.A.; Antunes, C.M.F.; Zicker, F.; Modabber, F. Failure of a Killed Leishmania amazonensis Vaccine against American Cutaneous Leishmaniasis in Colombia. Trans. R. Soc. Trop. Med. Hyg. 2005, 99, 593–598. [Google Scholar] [CrossRef]
  133. Palmieri, B.; Vadalà, M.; Roncati, L.; Garelli, A.; Scandone, F.; Bondi, M.; Cermelli, C. The Long-standing History of Corynebacterium Parvum, Immunity, and Viruses. J. Med. Virol. 2020, 92, 2429–2439. [Google Scholar] [CrossRef]
  134. Passero, L.F.D.; da Costa Bordon, M.L.A.; de Carvalho, A.K.; Martins, L.M.; Corbett, C.E.P.; Laurenti, M.D. Exacerbation of Leishmania (Viannia) Shawi Infection in BALB/c Mice after Immunization with Soluble Antigen from Amastigote Forms. APMIS 2010, 118, 973–981. [Google Scholar] [CrossRef] [PubMed]
  135. Reis, A.B.; Teixeira-Carvalho, A.; Giunchetti, R.C.; Guerra, L.L.; Carvalho, M.G.; Mayrink, W.; Genaro, O.; Corrêa-Oliveira, R.; Martins-Filho, O.A. Phenotypic Features of Circulating Leucocytes as Immunological Markers for Clinical Status and Bone Marrow Parasite Density in Dogs Naturally Infected by Leishmania Chagasi. Clin. Exp. Immunol. 2006, 146, 303–311. [Google Scholar] [CrossRef] [PubMed]
  136. Miles, A.P.; McClellan, H.A.; Rausch, K.M.; Zhu, D.; Whitmore, M.D.; Singh, S.; Martin, L.B.; Wu, Y.; Giersing, B.K.; Stowers, A.W.; et al. Montanide® ISA 720 Vaccines: Quality Control of Emulsions, Stability of Formulated Antigens, and Comparative Immunogenicity of Vaccine Formulations. Vaccine 2005, 23, 2530–2539. [Google Scholar] [CrossRef] [PubMed]
  137. Khalil, E.A.G.; Musa, A.M.; Modabber, F.; El-Hassan, A.M. Safety and Immunogenicity of a Candidate Vaccine for Visceral Leishmaniasis (Alum-Precipitated Autoclaved Leishmania Major + BCG) in Children: An Extended Phase II Study. Ann. Trop. Paediatr. 2006, 26, 357–361. [Google Scholar] [CrossRef]
  138. Bruhn, K.W.; Birnbaum, R.; Haskell, J.; Vanchinathan, V.; Greger, S.; Narayan, R.; Chang, P.-L.; Tran, T.A.; Hickerson, S.M.; Beverley, S.M.; et al. Killed but Metabolically Active Leishmania Infantum as a Novel Whole-Cell Vaccine for Visceral Leishmaniasis. Clin. Vaccine Immunol. 2012, 19, 490–498. [Google Scholar] [CrossRef]
  139. Pratti, J.E.S.; Ramos, T.D.; Pereira, J.C.; da Fonseca-Martins, A.M.; Maciel-Oliveira, D.; Oliveira-Silva, G.; de Mello, M.F.; Chaves, S.P.; Gomes, D.C.O.; Diaz, B.L.; et al. Efficacy of Intranasal LaAg Vaccine against Leishmania amazonensis Infection in Partially Resistant C57Bl/6 Mice. Parasit. Vectors 2016, 9, 534. [Google Scholar] [CrossRef]
  140. MALAFAIA, G.; SERAFIM, T.D.; SILVA, M.E.; PEDROSA, M.L.; REZENDE, S.A. Protein-energy Malnutrition Decreases Immune Response to Leishmania Chagasi Vaccine in BALB/c Mice. Parasite Immunol. 2009, 31, 41–49. [Google Scholar] [CrossRef]
  141. Ravindran, R.; Bhowmick, S.; Das, A.; Ali, N. Comparison of BCG, MPL and Cationic Liposome Adjuvant Systems in Leishmanial Antigen Vaccine Formulations against Murine Visceral Leishmaniasis. BMC Microbiol. 2010, 10, 181. [Google Scholar] [CrossRef]
  142. Trotta, T.; Fasanella, A.; Scaltrito, D.; Gradoni, L.; Mitolo, V.; Brandonisio, O.; Acquafredda, A.; Panaro, M.A. Comparison between Three Adjuvants for a Vaccine against Canine Leishmaniasis: In Vitro Evaluation of Macrophage Killing Ability. Comp. Immunol. Microbiol. Infect. Dis. 2010, 33, 175–182. [Google Scholar] [CrossRef]
  143. Coler, R.N.; Reed, S.G. Second-Generation Vaccines against Leishmaniasis. Trends Parasitol. 2005, 21, 244–249. [Google Scholar] [CrossRef]
  144. Stober, C.B.; Lange, U.G.; Mark, †; Roberts, T.M.; Alcami, A.; Blackwell, J.M. IL-10 from Regulatory T Cells Determines Vaccine Efficacy in Murine Leishmania Major Infection. J. Immunol. 2005, 175, 2517–2524. [Google Scholar] [CrossRef] [PubMed]
  145. Didwania, N.; Bhowmick, S.; Sabur, A.; Bhattacharya, A.; Ali, N. Evaluation of Leishmania Homolog of Activated C Kinase (LACK) of Leishmania Donovani in Comparison to Glycoprotein 63 as a Vaccine Candidate against Visceral Leishmaniasis. Microbiol. Spectr. 2025, 13. [Google Scholar] [CrossRef] [PubMed]
  146. Moreno, J.; Nieto, J.; Masina, S.; Cañavate, C.; Cruz, I.; Chicharro, C.; Carrillo, E.; Napp, S.; Reymond, C.; Kaye, P.M.; et al. Immunization with H1, HASPB1 and MML Leishmania Proteins in a Vaccine Trial against Experimental Canine Leishmaniasis. Vaccine 2007, 25, 5290–5300. [Google Scholar] [CrossRef] [PubMed]
  147. Bhowmick, S.; Ravindran, R.; Ali, N. Gp63 in Stable Cationic Liposomes Confers Sustained Vaccine Immunity to Susceptible BALB/c Mice Infected with Leishmania Donovani. Infect. Immun. 2008, 76, 1003–1015. [Google Scholar] [CrossRef]
  148. Fernández, L.; Carrillo, E.; Sánchez-Sampedro, L.; Sánchez, C.; Ibarra-Meneses, A.V.; Jimenez, M.A.; Almeida, V.d.A.; Esteban, M.; Moreno, J. Antigenicity of Leishmania-Activated C-Kinase Antigen (LACK) in Human Peripheral 181-Blood Mononuclear Cells, and Protective Effect of Prime-Boost Vaccination With PCI-Neo-LACK Plus Attenuated LACK-Expressing Vaccinia Viruses in Hamsters. Front. Immunol. 2018, 9, 843. [Google Scholar] [CrossRef]
  149. Olivier, M.; Atayde, V.D.; Isnard, A.; Hassani, K.; Shio, M.T. Leishmania Virulence Factors: Focus on the Metalloprotease GP63. Microbes Infect. 2012, 14, 1377–1389. [Google Scholar] [CrossRef]
  150. Kaur, T.; Sobti, R.C.; Kaur, S. Cocktail of Gp63 and Hsp70 Induces Protection against Leishmania Donovani in BALB/c Mice. Parasite Immunol. 2011, 33, 95–103. [Google Scholar] [CrossRef]
  151. Kaur, T.; Thakur, A.; Kaur, S. Protective Immunity Using MPL-A and Autoclaved Leishmania Donovani as Adjuvants along with a Cocktail Vaccine in Murine Model of Visceral Leishmaniasis. J. Parasit. Dis. 2013, 37, 231–239. [Google Scholar] [CrossRef]
  152. Bhowmick, S.; Ali, N. Identification of Novel Leishmania Donovani Antigens That Help Define Correlates of Vaccine-Mediated Protection in Visceral Leishmaniasis. PLoS ONE 2009, 4, e5820. [Google Scholar] [CrossRef]
  153. Nagill, R.; Kaur, T.; Joshi, J.; Kaur, S. Immunogenicity and Efficacy of Recombinant 78 KDa Antigen of Leishmania Donovani Formulated in Various Adjuvants against Murine Visceral Leishmaniasis. Asian Pac. J. Trop. Med. 2015, 8, 513–519. [Google Scholar] [CrossRef]
  154. Vakili, B.; Nezafat, N.; Zare, B.; Erfani, N.; Akbari, M.; Ghasemi, Y.; Rahbar, M.R.; Hatam, G.R. A New Multi-Epitope Peptide Vaccine Induces Immune Responses and Protection against Leishmania Infantum in BALB/c Mice. Med. Microbiol. Immunol. 2020, 209, 69–79. [Google Scholar] [CrossRef] [PubMed]
  155. Lage, D.P.; Ribeiro, P.A.F.; Dias, D.S.; Mendonça, D.V.C.; Ramos, F.F.; Carvalho, L.M.; de Oliveira, D.; Steiner, B.T.; Martins, V.T.; Perin, L.; et al. A Candidate Vaccine for Human Visceral Leishmaniasis Based on a Specific T Cell Epitope-Containing Chimeric Protein Protects Mice against Leishmania Infantum Infection. npj Vaccines 2020, 5, 1–13. [Google Scholar] [CrossRef] [PubMed]
  156. Rosa, R.; Marques, C.; Rodrigues, O.R.; Santos-Gomes, G.M. Immunization with Leishmania Infantum Released Proteins Confers Partial Protection against Parasite Infection with a Predominant Th1 Specific Immune Response. Vaccine 2007, 25, 4525–4532. [Google Scholar] [CrossRef] [PubMed]
  157. Passero, L.F.D.; Marques, C.; Vale-Gato, I.; Corbett, C.E.P.; Laurenti, M.D.; Santos-Gomes, G. Analysis of the Protective Potential of Antigens Released by Leishmania (Viannia) Shawi Promastigotes. Arch. Dermatol. Res. 2012, 304, 47–55. [Google Scholar] [CrossRef]
  158. Darrah, P.A.; Patel, D.T.; De Luca, P.M.; Lindsay, R.W.B.; Davey, D.F.; Flynn, B.J.; Hoff, S.T.; Andersen, P.; Reed, S.G.; Morris, S.L.; et al. Multifunctional TH1 Cells Define a Correlate of Vaccine-Mediated Protection against Leishmania Major. Nature Medicine 2007, 13, 843–850. [Google Scholar] [CrossRef]
  159. Fernández Cotrina, J.; Iniesta, V.; Monroy, I.; Baz, V.; Hugnet, C.; Marañon, F.; Fabra, M.; Gómez-Nieto, L.C.; Alonso, C. A Large-Scale Field Randomized Trial Demonstrates Safety and Efficacy of the Vaccine LetiFend® against Canine Leishmaniosis. Vaccine 2018, 36, 1972–1982. [Google Scholar] [CrossRef]
  160. Grimaldi, G.; Teva, A.; Dos-Santos, C.B.; Santos, F.N.; Pinto, I.D.S.; Fux, B.; Leite, G.R.; Falqueto, A. Field Trial of Efficacy of the Leish-Tec® Vaccine against Canine Leishmaniasis Caused by Leishmania Infantum in an Endemic Area with High Transmission Rates. PLoS ONE 2017, 12, e0185438. [Google Scholar] [CrossRef]
  161. Oliva, G.; Nieto, J.; Foglia Manzillo, V.; Cappiello, S.; Fiorentino, E.; Di Muccio, T.; Scalone, A.; Moreno, J.; Chicharro, C.; Carrillo, E.; et al. A Randomised, Double-Blind, Controlled Efficacy Trial of the LiESP/QA-21 Vaccine in Naïve Dogs Exposed to Two Leishmania Infantum Transmission Seasons. PLoS Negl. Trop. Dis. 2014, 8, e3213. [Google Scholar] [CrossRef]
  162. Nogueira, F.S.; Moreira, M.A.B.; Borja-Cabrera, G.P.; Santos, F.N.; Menz, I.; Parra, L.E.; Xu, Z.; Chu, H.J.; Palatnik-de-Sousa, C.B.; Luvizotto, M.C.R. Leishmune® Vaccine Blocks the Transmission of Canine Visceral Leishmaniasis: Absence of Leishmania Parasites in Blood, Skin and Lymph Nodes of Vaccinated Exposed Dogs. Vaccine 2005, 23, 4805–4810. [Google Scholar] [CrossRef]
  163. de A. Luna, E.J.; de S.L. da C. Campos, S.R. O Desenvolvimento de Vacinas Contra as Doenças Tropicais Negligenciadas. Cad. Saude Publica 2020, 36. [Google Scholar] [CrossRef]
  164. Palatnik-de-Sousa, C.B.; Paraguai-de-Souza, E.; Gomes, E.M.; Borojevic, R. Experimental Murine Leishmania Donovani Infection: Immunoprotection by the Fucose-Mannose Ligand (FML). Braz. J. Med. Biol. Res. = Revista brasileira de pesquisas medicas e biologica 1994, 27, 547–551. [Google Scholar]
  165. Marcondes, M.; Ikeda, F.A.; Vieira, R.F.C.; Day, M.J.; Lima, V.M.F.; Rossi, C.N.; Perri, S.H.V.; Biondo, A.W. Temporal IgG Subclasses Response in Dogs Following Vaccination against Leishmania with Leishmune®. Vet. Parasitol. 2011, 181, 153–159. [Google Scholar] [CrossRef] [PubMed]
  166. Fernandes, C.B.; Junior, J.T.M.; De Jesus, C.; Da Silva Souza, B.M.P.; Larangeira, D.F.; Fraga, D.B.M.; Tavares Veras, P.S.; Barrouin-Melo, S.M. Comparison of Two Commercial Vaccines against Visceral Leishmaniasis in Dogs from Endemic Areas: IgG, and Subclasses, Parasitism, and Parasite Transmission by Xenodiagnosis. Vaccine 2014, 32, 1287–1295. [Google Scholar] [CrossRef]
  167. Regina-Silva, S.; Feres, A.M.L.T.; França-Silva, J.C.; Dias, E.S.; Michalsky, É.M.; de Andrade, H.M.; Coelho, E.A.F.; Ribeiro, G.M.; Fernandes, A.P.; Machado-Coelho, G.L.L. Field Randomized Trial to Evaluate the Efficacy of the Leish-Tec® Vaccine against Canine Visceral Leishmaniasis in an Endemic Area of Brazil. Vaccine 2016, 34, 2233–2239. [Google Scholar] [CrossRef] [PubMed]
  168. Martiniano de Pádua, J.A.; Ribeiro, D.; de Aguilar, V.F.F.; Melo, T.F.; Bueno, L.L.; Grossi de Oliveira, A.L.; Fujiwara, R.T.; Keller, K.M. Overview of Commercial Vaccines Against Canine Visceral Leishmaniasis: Current Landscape and Future Directions. Pathogens 2025, 14, 970. [Google Scholar] [CrossRef]
  169. Soto, M.; Requena, J.M.; Quijada, L.; Alonso, C. Multicomponent Chimeric Antigen for Serodiagnosis of Canine Visceral Leishmaniasis. J. Clin. Microbiol. 1998, 36, 58–63. [Google Scholar] [CrossRef]
  170. Parody, N.; Soto, M.; Requena, J.M.; Alonso, C. Adjuvant Guided Polarization of the Immune Humoral Response against a Protective Multicomponent Antigenic Protein (Q) from Leishmania Infantum. A CpG + Q Mix Protects Balb/c Mice from Infection. Parasite Immunol. 2004, 26, 283–293. [Google Scholar] [CrossRef]
  171. Molano, I.; Alonso, M.G.; Mirón, C.; Redondo, E.; Requena, J.M.; Soto, M.; Nieto, C.G.; Alonso, C. A Leishmania Infantum Multi-Component Antigenic Protein Mixed with Live BCG Confers Protection to Dogs Experimentally Infected with L. Infantum. Vet. Immunol. Immunopathol. 2003, 92, 1–13. [Google Scholar] [CrossRef]
  172. Carcelén, J.; Iniesta, V.; Fernández-Cotrina, J.; Serrano, F.; Parejo, J.C.; Corraliza, I.; Gallardo-Soler, A.; Marañón, F.; Soto, M.; Alonso, C.; et al. The Chimerical Multi-Component Q Protein from Leishmania in the Absence of Adjuvant Protects Dogs against an Experimental Leishmania Infantum Infection. Vaccine 2009, 27, 5964–5973. [Google Scholar] [CrossRef]
  173. Coler, R.N.; Skeiky, Y.A.W.; Bernards, K.; Greeson, K.; Carter, D.; Cornellison, C.D.; Modabber, F.; Campos-Neto, A.; Reed, S.G. Immunization with a Polyprotein Vaccine Consisting of the T-Cell Antigens Thiol-Specific Antioxidant, Leishmania Major. Stress-Inducible Protein 1, and Leishmania Elongation Initiation Factor Protects against Leishmaniasis. Infect. Immun. 2002, 70, 4215–4225. [Google Scholar] [CrossRef]
  174. Lemesre, J.L.; Holzmuller, P.; Cavaleyra, M.; Gonçalves, R.B.; Hottin, G.; Papierok, G. Protection against Experimental Visceral Leishmaniasis Infection in Dogs Immunized with Purified Excreted Secreted Antigens of Leishmania Infantum Promastigotes. Vaccine 2005, 23, 2825–2840. [Google Scholar] [CrossRef] [PubMed]
  175. Lemesre, J.L.; Holzmuller, P.; Gonçalves, R.B.; Bourdoiseau, G.; Hugnet, C.; Cavaleyra, M.; Papierok, G. Long-Lasting Protection against Canine Visceral Leishmaniasis Using the LiESAp-MDP Vaccine in Endemic Areas of France: Double-Blind Randomised Efficacy Field Trial. Vaccine 2007, 25, 4223–4234. [Google Scholar] [CrossRef] [PubMed]
  176. Bourdoiseau, G.; Hugnet, C.; Gonçalves, R.B.; Vézilier, F.; Petit-Didier, E.; Papierok, G.; Lemesre, J.L. Effective Humoral and Cellular Immunoprotective Responses in Li ESAp-MDP Vaccinated Protected Dogs. Vet. Immunol. Immunopathol. 2009, 128, 71–78. [Google Scholar] [CrossRef] [PubMed]
  177. Salay, G.; Dorta, M.L.; Santos, N.M.; Mortara, R.A.; Brodskyn, C.; Oliveira, C.I.; Barbiéri, C.L.; Rodrigues, M.M. Testing of Four Leishmania Vaccine Candidates in a Mouse Model of Infection with Leishmania (Viannia) Braziliensis, the Main Causative Agent of Cutaneous Leishmaniasis in the New World. Clin. Vaccine Immunol. 2007, 14, 1173–1181. [Google Scholar] [CrossRef]
  178. Chamakh-Ayari, R.; Bras-Gonçalves, R.; Bahi-Jaber, N.; Petitdidier, E.; Markikou-Ouni, W.; Aoun, K.; Moreno, J.; Carrillo, E.; Salotra, P.; Kaushal, H.; et al. In Vitro Evaluation of a Soluble Leishmania Promastigote Surface Antigen as a Potential Vaccine Candidate against Human Leishmaniasis. PLoS ONE 2014, 9, e92708. [Google Scholar] [CrossRef]
  179. de Matos Guedes, H.L.; Pinheiro, R.O.; Chaves, S.P.; De-Simone, S.G.; Rossi-Bergmann, B. Serine Proteases of Leishmania amazonensis as Immunomodulatory and Disease-Aggravating Components of the Crude LaAg Vaccine. Vaccine 2010, 28, 5491–5496. [Google Scholar] [CrossRef]
  180. Kushawaha, P.K.; Gupta, R.; Tripathi, C.D.P.; Sundar, S.; Dube, A. Evaluation of Leishmania Donovani Protein Disulfide Isomerase as a Potential Immunogenic Protein/Vaccine Candidate against Visceral Leishmaniasis. PLoS ONE 2012, 7, e35670. [Google Scholar] [CrossRef]
  181. Kushawaha, P.K.; Gupta, R.; Tripathi, C.D.P.; Khare, P.; Jaiswal, A.K.; Sundar, S.; Dube, A. Leishmania Donovani Triose Phosphate Isomerase: A Potential Vaccine Target against Visceral Leishmaniasis. PLoS ONE 2012, 7, e45766. [Google Scholar] [CrossRef]
  182. Vélez, I.D.; Gilchrist, K.; Martínez, S.; Ramírez-Pineda, J.R.; Ashman, J.A.; Alves, F.P.; Coler, R.N.; Bogatzki, L.Y.; Kahn, S.J.; Beckmann, A.M.; et al. Safety and Immunogenicity of a Defined Vaccine for the Prevention of Cutaneous Leishmaniasis. Vaccine 2009, 28, 329–337. [Google Scholar] [CrossRef]
  183. Chakravarty, J.; Kumar, S.; Trivedi, S.; Rai, V.K.; Singh, A.; Ashman, J.A.; Laughlin, E.M.; Coler, R.N.; Kahn, S.J.; Beckmann, A.M.; et al. A Clinical Trial to Evaluate the Safety and Immunogenicity of the LEISH-F1+MPL-SE Vaccine for Use in the Prevention of Visceral Leishmaniasis. Vaccine 2011, 29, 3531–3537. [Google Scholar] [CrossRef]
  184. de Amorim, I.F.G.; Freitas, E.; Alves, C.F.; Tafuri, W.L.; Melo, M.N.; Michalick, M.S.M.; da Costa-Val, A.P. Humoral Immunological Profile and Parasitological Statuses of Leishmune® Vaccinated and Visceral Leishmaniasis Infected Dogs from an Endemic Area. Vet. Parasitol. 2010, 173, 55–63. [Google Scholar] [CrossRef] [PubMed]
  185. Dias, D.S.; Ribeiro, P.A.F.; Martins, V.T.; Lage, D.P.; Ramos, F.F.; Dias, A.L.T.; Rodrigues, M.R.; Portela, Á.S.B.; Costa, L.E.; Caligiorne, R.B.; et al. Recombinant Prohibitin Protein of Leishmania Infantum Acts as a Vaccine Candidate and Diagnostic Marker against Visceral Leishmaniasis. Cell Immunol. 2018, 323, 59–69. [Google Scholar] [CrossRef] [PubMed]
  186. de Oliveira Emerick, S.; Vieira de Carvalho, T.; Meirelles Miranda, B.; Carneiro da Silva, A.; Viana Fialho Martins, T.; Licursi de Oliveira, L.; de Almeida Marques-da-Silva, E. Lipophosphoglycan-3 Protein from Leishmania Infantum Chagasi plus Saponin Adjuvant: A New Promising Vaccine against Visceral Leishmaniasis. Vaccine 2021, 39, 282–291. [Google Scholar] [CrossRef] [PubMed]
  187. Iniguez, E.; Schocker, N.S.; Subramaniam, K.; Portillo, S.; Montoya, A.L.; Al-Salem, W.S.; Torres, C.L.; Rodriguez, F.; Moreira, O.C.; Acosta-Serrano, A.; et al. An α-Gal-Containing Neoglycoprotein-Based Vaccine Partially Protects against Murine Cutaneous Leishmaniasis Caused by Leishmania Major. PLoS Negl. Trop. Dis. 2017, 11, e0006039. [Google Scholar] [CrossRef]
  188. Malone, R.W.; Felgner, P.L.; Verma, I.M. Cationic Liposome-Mediated RNA Transfection. Proc. Natl. Acad. Sci. USA 1989, 86, 6077–6081. [Google Scholar] [CrossRef]
  189. Neeli, P.; Chai, D.; Roy, D.; Prajapati, S.; Bonam, S.R. DNA Vaccines in the Post-MRNA Era: Engineering, Applications, and Emerging Innovations. Int. J. Mol. Sci. 2025, 26, 8716. [Google Scholar] [CrossRef]
  190. Neefjes, J.; Jongsma, M.L.M.; Paul, P.; Bakke, O. Towards a Systems Understanding of MHC Class I and MHC Class II Antigen Presentation. Nat. Rev. Immunol. 2011, 11, 823–836. [Google Scholar] [CrossRef]
  191. Liu, M.A. DNA Vaccines: A Review. J. Intern. Med. 2003, 253, 402–410. [Google Scholar] [CrossRef]
  192. Zhou, W.; Jiang, L.; Liao, S.; Wu, F.; Yang, G.; Hou, L.; Liu, L.; Pan, X.; Jia, W.; Zhang, Y. Vaccines’ New Era-RNA Vaccine. Viruses 2023, 15, 1760. [Google Scholar] [CrossRef]
  193. Xu, D.; Liew, F.Y. Protection against Leishmaniasis by Injection of DNA Encoding a Major Surface Glycoprotein, Gp63, of L. Major. Immunology 1995, 84, 173–176. [Google Scholar]
  194. Saltzman, W.M.; Shen, H.; Brandsma, J.L. DNA Vaccines: Methods and Protocols; Humana Press: Totowa, NJ, USA, 2006; ISBN 1588294846. [Google Scholar]
  195. Mazumder, S.; Maji, M.; Das, A.; Ali, N. Potency, Efficacy and Durability of DNA/DNA, DNA/Protein and Protein/Protein Based Vaccination Using Gp63 Against Leishmania Donovani in BALB/c Mice. PLoS ONE 2011, 6, e14644. [Google Scholar] [CrossRef]
  196. Sachdeva, R.; Banerjea, A.C.; Malla, N.; Dubey, M.L. Immunogenicity and Efficacy of Single Antigen Gp63, Polytope and PolytopeHSP70 DNA Vaccines against Visceral Leishmaniasis in Experimental Mouse Model. PLoS ONE 2009, 4, e7880. [Google Scholar] [CrossRef] [PubMed]
  197. Ali, S.A.; Rezvan, H.; Mcardle, S.E.; Khodadadi, A.; Asteal, F.A.; Rees, R.C. CTL Responses to Leishmania Mexicana Gp63-cDNA Vaccine in a Murine Model. Parasite Immunol. 2009, 31, 373–383. [Google Scholar] [CrossRef] [PubMed]
  198. Zhang, J.; He, J.; Liao, X.; Xiao, Y.; Liang, C.; Zhou, Q.; Chen, H.; Zheng, Z.; Qin, H.; Chen, D.; et al. Development of Dominant Epitope-Based Vaccines Encoding Gp63, Kmp-11 and Amastin against Visceral Leishmaniasis. Immunobiology 2021, 226, 152085. [Google Scholar] [CrossRef] [PubMed]
  199. Kelly, B.L.; Stetson, D.B.; Locksley, R.M. Leishmania Major LACK Antigen Is Required for Efficient Vertebrate Parasitization. J. Exp. Med. 2003, 198, 1689–1698. [Google Scholar] [CrossRef]
  200. Gurunathan, S.; Sacks, D.L.; Brown, D.R.; Reiner, S.L.; Charest, H.; Glaichenhaus, N.; Seder, R.A. Vaccination with DNA Encoding the Immunodominant LACK Parasite Antigen Confers Protective Immunity to Mice Infected with Leishmania Major. J. Exp. Med. 1997, 186, 1137–1147. [Google Scholar] [CrossRef]
  201. Gurunathan, S.; Stobie, L.; Prussin, C.; Sacks, D.L.; Glaichenhaus, N.; Fowell, D.J.; Locksley, R.M.; Chang, J.T.; Wu, C.-Y.; Seder, R.A. Requirements for the Maintenance of Th1 Immunity In Vivo Following DNA Vaccination: A Potential Immunoregulatory Role for CD8+ T Cells. J. Immunol. 2000, 165, 915–924. [Google Scholar] [CrossRef]
  202. Melby, P.C.; Yang, J.; Zhao, W.; Perez, L.E.; Cheng, J. Leishmania donovani P36(LACK) DNA Vaccine Is Highly Immunogenic but Not Protective against Experimental Visceral Leishmaniasis. Infect. Immun. 2001, 69, 4719–4725. [Google Scholar] [CrossRef]
  203. Marques-da-Silva, E.A.; Coelho, E.A.F.; Gomes, D.C.O.; Vilela, M.C.; Masioli, C.Z.; Tavares, C.A.P.; Fernandes, A.P.; Afonso, L.C.C.; Rezende, S.A. Intramuscular Immunization with P36(LACK) DNA Vaccine Induces IFN-γ Production but Does Not Protect BALB/c Mice against Leishmania Chagasi Intravenous Challenge. Parasitol. Res. 2005, 98, 67–74. [Google Scholar] [CrossRef]
  204. Pinto, E.F.; Pinheiro, R.O.; Rayol, A.; Larraga, V.; Rossi-Bergmann, B. Intranasal Vaccination against Cutaneous Leishmaniasis with a Particulated Leishmanial Antigen or DNA Encoding LACK. Infect. Immun. 2004, 72, 4521–4527. [Google Scholar] [CrossRef]
  205. Gomes, D.C.d.O.; Pinto, E.F.; de Melo, L.D.B.; Lima, W.P.; Larraga, V.; Lopes, U.G.; Rossi-Bergmann, B. Intranasal Delivery of Naked DNA Encoding the LACK Antigen Leads to Protective Immunity against Visceral Leishmaniasis in Mice. Vaccine 2007, 25, 2168–2172. [Google Scholar] [CrossRef] [PubMed]
  206. De Oliveira Gomes, D.C.; Da Silva Costa Souza, B.L.; De Matos Guedes, H.L.; Lopes, U.G.; Rossi-Bergmann, B. Intranasal Immunization with LACK-DNA Promotes Protective Immunity in Hamsters Challenged with Leishmania Chagasi. Parasitology 2011, 138, 1892–1897. [Google Scholar] [CrossRef] [PubMed]
  207. Costa Souza, B.L.S.; Pinto, E.F.; Bezerra, I.P.S.; Gomes, D.C.O.; Martinez, A.M.B.; Ré, M.I.; de Matos Guedes, H.L.; Rossi-Bergmann, B. Crosslinked Chitosan Microparticles as a Safe and Efficient DNA Carrier for Intranasal Vaccination against Cutaneous Leishmaniasis. Vaccine X 2023, 15, 100403. [Google Scholar] [CrossRef] [PubMed]
  208. Gomes, D.C.O.; Souza, B.L.d.S.C.; Schwedersky, R.P.; Covre, L.P.; de Matos Guedes, H.L.; Lopes, U.G.; Ré, M.I.; Rossi-Bergmann, B. Intranasal Immunization with Chitosan Microparticles Enhances LACK-DNA Vaccine Protection and Induces Specific Long-Lasting Immunity against Visceral Leishmaniasis. Microbes Infect. 2022, 24, 104884. [Google Scholar] [CrossRef]
  209. Pérez-Jiménez, E.; Kochan, G.; Gherardi, M.M.; Esteban, M. MVA-LACK as a Safe and Efficient Vector for Vaccination against Leishmaniasis. Microbes Infect. 2006, 8, 810–822. [Google Scholar] [CrossRef]
  210. Ramos, I.; Alonso, A.; Peris, A.; Marcen, J.M.; Abengozar, M.A.; Alcolea, P.J.; Castillo, J.A.; Larraga, V. Antibiotic Resistance Free Plasmid DNA Expressing LACK Protein Leads towards a Protective Th1 Response against Leishmania Infantum Infection. Vaccine 2009, 27, 6695–6703. [Google Scholar] [CrossRef]
  211. Alonso, A.; Alcolea, P.J.; Larraga, J.; Peris, M.P.; Esteban, A.; Cortés, A.; Ruiz-García, S.; Castillo, J.A.; Larraga, V. A Non-Replicative Antibiotic Resistance-Free DNA Vaccine Delivered by the Intranasal Route Protects against Canine Leishmaniasis. Front Immunol 2023, 14. [Google Scholar] [CrossRef]
  212. Rodríguez-Almonacid, C.C.; Kellogg, M.K.; Karamyshev, A.L.; Karamysheva, Z.N. Ribosome Specialization in Protozoa Parasites. Int. J. Mol. Sci. 2023, 24, 7484. [Google Scholar] [CrossRef]
  213. Soto, M.; Alonso, C.; Requena, J.M. The Leishmania Infantum Acidic Ribosomal Protein LiP2a Induces a Prominent Humoral Response in Vivo and Stimulates Cell Proliferation in Vitro and Interferon-Gamma (IFN-γ) Production by Murine Splenocytes. Clin. Exp. Immunol. 2008, 122, 212–218. [Google Scholar] [CrossRef]
  214. Requena, J.M.; Alonso, C.; Soto, M. Evolutionarily Conserved Proteins as Prominent Immunogens during Leishmania Infections. Parasitol. Today 2000, 16, 246–250. [Google Scholar] [CrossRef]
  215. Iborra, S.; Soto, M.; Carrión, J.; Nieto, A.; Fernández, E.; Alonso, C.; Requena, J.M. The Leishmania Infantum Acidic Ribosomal Protein P0 Administered as a DNA Vaccine Confers Protective Immunity to Leishmania Major Infection in BALB/c Mice. Infect. Immun. 2003, 71, 6562–6572. [Google Scholar] [CrossRef] [PubMed]
  216. Pereira, L.; Abbehusen, M.; Teixeira, C.; Cunha, J.; Nascimento, I.P.; Fukutani, K.; dos-Santos, W.; Barral, A.; de Oliveira, C.I.; Barral-Netto, M.; et al. Vaccination with Leishmania Infantum Acidic Ribosomal P0 but Not with Nucleosomal Histones Proteins Controls Leishmania Infantum Infection in Hamsters. PLoS Negl. Trop. Dis. 2015, 9, e0003490. [Google Scholar] [CrossRef] [PubMed]
  217. Reverte, M.; Eren, R.O.; Jha, B.; Desponds, C.; Snäkä, T.; Prevel, F.; Isorce, N.; Lye, L.-F.; Owens, K.L.; Gazos Lopes, U.; et al. The Antioxidant Response Favors Leishmania Parasites Survival, Limits Inflammation and Reprograms the Host Cell Metabolism. PLoS Pathog. 2021, 17, e1009422. [Google Scholar] [CrossRef] [PubMed]
  218. Stober, C.B.; Lange, U.G.; Roberts, M.T.M.; Alcami, A.; Blackwell, J.M. Heterologous Priming-Boosting with DNA and Modified Vaccinia Virus Ankara Expressing Tryparedoxin Peroxidase Promotes Long-Term Memory against Leishmania Major in Susceptible BALB/c Mice. Infect. Immun. 2007, 75, 852–860. [Google Scholar] [CrossRef]
  219. Jayakumar, A.; Castilho, T.M.; Park, E.; Goldsmith-Pestana, K.; Blackwell, J.M.; McMahon-Pratt, D. TLR1/2 Activation during Heterologous Prime-Boost Vaccination (DNA-MVA) Enhances CD8+ T Cell Responses Providing Protection against Leishmania (Viannia). PLoS Negl. Trop. Dis. 2011, 5, e1204. [Google Scholar] [CrossRef]
  220. Abreu, I.A.; Cabelli, D.E. Superoxide dismutases-a review of the metal-associated mechanistic variations. Biochim. Biophys. Acta 2010, 1804, 263–274. [Google Scholar] [CrossRef]
  221. Plewes, K.A.; Barr, S.D.; Gedamu, L. Iron Superoxide Dismutases Targeted to the Glycosomes of Leishmania Chagasi Are Important for Survival. Infect. Immun. 2003, 71, 5910–5920. [Google Scholar] [CrossRef]
  222. Getachew, F.; Gedamu, L. Leishmania Donovani Iron Superoxide Dismutase A Is Targeted to the Mitochondria by Its N-Terminal Positively Charged Amino Acids. Mol. Biochem. Parasitol. 2007, 154, 62–69. [Google Scholar] [CrossRef]
  223. Dufernez, F.; Yernaux, C.; Gerbod, D.; Noël, C.; Chauvenet, M.; Wintjens, R.; Edgcomb, V.P.; Capron, M.; Opperdoes, F.R.; Viscogliosi, E. The Presence of Four Iron-Containing Superoxide Dismutase Isozymes in Trypanosomatidae: Characterization, Subcellular Localization, and Phylogenetic Origin in Trypanosoma Brucei. Free Radic. Biol. Med. 2006, 40, 210–225. [Google Scholar] [CrossRef]
  224. Bayih, A.G.; Daifalla, N.S.; Gedamu, L. Immune Response and Protective Efficacy of a Heterologous DNA-Protein Immunization with Leishmania Superoxide Dismutase B1. Biomed. Res. Int. 2017, 2017, 2145386. [Google Scholar] [CrossRef]
  225. Campos, B.L.S.; Silva, T.N.; Ribeiro, S.P.; Carvalho, K.I.L.; KallÁs, E.G.; Laurenti, M.D.; Passero, L.F.D. Analysis of Iron Superoxide Dismutase-encoding DNA Vaccine on the Evolution of the Leishmania amazonensis Experimental Infection. Parasite Immunol. 2015, 37, 407–416. [Google Scholar] [CrossRef] [PubMed]
  226. Ghaffarifar, F.; Jorjani, O.; Sharifi, Z.; Dalimi, A.; Hassan, Z.M.; Tabatabaie, F.; Khoshzaban, F.; Hezarjaribi, H.Z. Enhancement of Immune Response Induced by DNA Vaccine Cocktail Expressing Complete LACK and TSA Genes against Leishmania Major. APMIS 2013, 121, 290–298. [Google Scholar] [CrossRef] [PubMed]
  227. Maspi, N.; Ghaffarifar, F.; Sharifi, Z.; Dalimi, A.; Khademi, S.Z. DNA Vaccination with a Plasmid Encoding LACK-TSA Fusion against Leishmania Major Infection in BALB/c Mice. Malays. J. Pathol. 2017, 39, 267–275. [Google Scholar] [PubMed]
  228. Campos-Neto, A.; Webb, J.R.; Greeson, K.; Coler, R.N.; Skeiky, Y.A.W.; Reed, S.G. Vaccination with Plasmid DNA Encoding TSA/LmSTI1 Leishmanial Fusion Proteins Confers Protection against Leishmania Major Infection in Susceptible BALB/c Mice. Infect. Immun. 2002, 70, 2828–2836. [Google Scholar] [CrossRef]
  229. Saldarriaga, O.A.; Travi, B.L.; Park, W.; Perez, L.E.; Melby, P.C. Immunogenicity of a Multicomponent DNA Vaccine against Visceral Leishmaniasis in Dogs. Vaccine 2006, 24, 1928–1940. [Google Scholar] [CrossRef]
  230. Das, S.; Freier, A.; Boussoffara, T.; Das, S.; Oswald, D.; Losch, F.O.; Selka, M.; Sacerdoti-Sierra, N.; Schönian, G.; Wiesmüller, K.-H.; et al. Modular Multiantigen T Cell Epitope–Enriched DNA Vaccine Against Human Leishmaniasis. Sci. Transl. Med. 2014, 6. [Google Scholar] [CrossRef]
  231. Rodríguez-Cortés, A.; Ojeda, A.; López-Fuertes, L.; Timón, M.; Altet, L.; Solano-Gallego, L.; Sánchez-Robert, E.; Francino, O.; Alberola, J. Vaccination with Plasmid DNA Encoding KMPII, TRYP, LACK and GP63 Does Not Protect Dogs against Leishmania Infantum Experimental Challenge. Vaccine 2007, 25, 7962–7971. [Google Scholar] [CrossRef]
  232. Webb, J.R.; Campos-Neto, A.; Ovendale, P.J.; Martin, T.I.; Stromberg, E.J.; Badaro, R.; Reed, S.G. Human and Murine Immune Responses to a Novel Leishmania Major Recombinant Protein Encoded by Members of a Multicopy Gene Family. Infect. Immun. 1998, 66, 3279–3289. [Google Scholar] [CrossRef]
  233. Goto, Y.; Bhatia, A.; Raman, V.S.; Liang, H.; Mohamath, R.; Picone, A.F.; Vidal, S.E.Z.; Vedvick, T.S.; Howard, R.F.; Reed, S.G. KSAC, the First Defined Polyprotein Vaccine Candidate for Visceral Leishmaniasis. Clin. Vaccine Immunol. 2011, 18, 1118–1124. [Google Scholar] [CrossRef]
Figure 1. Diagram showing in (A) nonspecific inflammatory response to infection by parasites of the genus Leishmania, involving innate immune cells such as dendritic cells (DC), macrophages (M), neutrophils (N), natural killer cells (NT), and the complement system proteins (C). In (B) the activation of specific immunity in regional immune organs such as lymph nodes (LN) can be observed with antigen presentation by the MHC and specific lymphocyte stimulation, such as: T helper 1 (Th1) with production of pro-inflammatory cytokines (IFN-γ, IL-2, IL-12, TNF-α), T helper 2 with production of anti-inflammatory cytokines (IL-4, IL-13), T regulatory cells with production of regulatory cytokines (IL-10, TGF-β) and T cytotoxic (TCL) with cytotoxic action or IFN-γ production. B lymphocytes (B) can also be activated for antibody production.
Figure 1. Diagram showing in (A) nonspecific inflammatory response to infection by parasites of the genus Leishmania, involving innate immune cells such as dendritic cells (DC), macrophages (M), neutrophils (N), natural killer cells (NT), and the complement system proteins (C). In (B) the activation of specific immunity in regional immune organs such as lymph nodes (LN) can be observed with antigen presentation by the MHC and specific lymphocyte stimulation, such as: T helper 1 (Th1) with production of pro-inflammatory cytokines (IFN-γ, IL-2, IL-12, TNF-α), T helper 2 with production of anti-inflammatory cytokines (IL-4, IL-13), T regulatory cells with production of regulatory cytokines (IL-10, TGF-β) and T cytotoxic (TCL) with cytotoxic action or IFN-γ production. B lymphocytes (B) can also be activated for antibody production.
Vaccines 14 00054 g001
Figure 2. Alum’s mechanism of action. Antigens and adjuvants are slowly released into the tissue (1), recruiting innate immune cells to the site of immunization (2). Dendritic cells phagocytosis and process the antigen, presenting it to CD4+ T lymphocytes through MHCII molecules (3). At the same time, the adjuvant stimulates the endogenous immune response through the activation of the NLRP3 protein (4), which binds to ASC and pro-caspase 1, and caspase 1, assembling the NLRP3 inflammasome (5), which promotes the secretion of pro-inflammatory cytokines such as IL-1β, IL-18, and IL-33 (6). These cytokines drive the differentiation of CD4+ T cells into the Th2 subset (7), stimulating the production of antibodies by B cells (8).
Figure 2. Alum’s mechanism of action. Antigens and adjuvants are slowly released into the tissue (1), recruiting innate immune cells to the site of immunization (2). Dendritic cells phagocytosis and process the antigen, presenting it to CD4+ T lymphocytes through MHCII molecules (3). At the same time, the adjuvant stimulates the endogenous immune response through the activation of the NLRP3 protein (4), which binds to ASC and pro-caspase 1, and caspase 1, assembling the NLRP3 inflammasome (5), which promotes the secretion of pro-inflammatory cytokines such as IL-1β, IL-18, and IL-33 (6). These cytokines drive the differentiation of CD4+ T cells into the Th2 subset (7), stimulating the production of antibodies by B cells (8).
Vaccines 14 00054 g002
Figure 3. The mechanism of action of CFA (A) begins with the deposition of heat-killed mycobacteria in a w/o emulsion (1), which, when injected, recruits immune cells to the site of immunization (2). The antigen and adjuvant are processed by APCs, which release pro-inflammatory cytokines (IL-6, TNF-α, and IL-12), recruiting NK cells (3). These cells produce IL-12 and IFN-γ, stimulating the differentiation of Th1 lymphocytes (4). The w/o emulsion with paraffin and mycobacteria prolongs the Th1 response, leading to DTH (5), inflammatory and toxic effects to the host (6). In IFA (B), deposition of the adjuvant with antigen recruits immune cells (1), which are processed by APCs (2). The absence of mycobacteria in the adjuvant results in the production of cytokines IL-1β and IL-18, stimulating CD4+ T lymphocytes (3). These cells produce IL-4, IL-1β, and IL-18, promoting the maturation of the Th2 cell subset (4). Th2 cell subset, in turn, produces IL-4, IL-5, and IL-10 (5), activating B cells and inducing antibody production (6), establishing the humoral response.
Figure 3. The mechanism of action of CFA (A) begins with the deposition of heat-killed mycobacteria in a w/o emulsion (1), which, when injected, recruits immune cells to the site of immunization (2). The antigen and adjuvant are processed by APCs, which release pro-inflammatory cytokines (IL-6, TNF-α, and IL-12), recruiting NK cells (3). These cells produce IL-12 and IFN-γ, stimulating the differentiation of Th1 lymphocytes (4). The w/o emulsion with paraffin and mycobacteria prolongs the Th1 response, leading to DTH (5), inflammatory and toxic effects to the host (6). In IFA (B), deposition of the adjuvant with antigen recruits immune cells (1), which are processed by APCs (2). The absence of mycobacteria in the adjuvant results in the production of cytokines IL-1β and IL-18, stimulating CD4+ T lymphocytes (3). These cells produce IL-4, IL-1β, and IL-18, promoting the maturation of the Th2 cell subset (4). Th2 cell subset, in turn, produces IL-4, IL-5, and IL-10 (5), activating B cells and inducing antibody production (6), establishing the humoral response.
Vaccines 14 00054 g003
Figure 4. BCG adjuvant leads to the activation of macrophages, neutrophils, and dendritic cells (1), presenting antigens to CD4+ and CD8+ T lymphocytes (2), which lead to an adaptive immune response. CD4+ T lymphocytes stimulate B lymphocytes (3), which produce memory and plasma cells (4). In addition, BCG adjuvant induces metabolic reprogramming that increases glycolysis, by the signalization of NOD2 (5), providing the energy needed for cellular activation (6). Concomitantly, it induces epigenetic reprogramming in innate immune cells through the opening of chromatin in promoter regions (7), leading to the high expression of pro-inflammatory cytokines (8).
Figure 4. BCG adjuvant leads to the activation of macrophages, neutrophils, and dendritic cells (1), presenting antigens to CD4+ and CD8+ T lymphocytes (2), which lead to an adaptive immune response. CD4+ T lymphocytes stimulate B lymphocytes (3), which produce memory and plasma cells (4). In addition, BCG adjuvant induces metabolic reprogramming that increases glycolysis, by the signalization of NOD2 (5), providing the energy needed for cellular activation (6). Concomitantly, it induces epigenetic reprogramming in innate immune cells through the opening of chromatin in promoter regions (7), leading to the high expression of pro-inflammatory cytokines (8).
Vaccines 14 00054 g004
Figure 5. MPL or GLA is injected as a vaccine adjuvant (1). The adjuvant particles stimulate TLR-4 expressed on the cell membrane (2), which is activated by the coreceptors CD14 and MD-2 (3), recruiting the MyD88 protein (4). The signaling cascade activates NF-kB (5), initiating the expression of pro-inflammatory cytokines such as IL-1β and IL-6, and TNF-α (6). The production of pro-inflammatory cytokines and the action of CD14 stimulate the processing of antigens and their presentation to the MHC (7), which results in the maturation of APCs, as well as the activation of IFN-γ-producing CD4+ T lymphocytes (8), which leads to polarization towards Th1 (9).
Figure 5. MPL or GLA is injected as a vaccine adjuvant (1). The adjuvant particles stimulate TLR-4 expressed on the cell membrane (2), which is activated by the coreceptors CD14 and MD-2 (3), recruiting the MyD88 protein (4). The signaling cascade activates NF-kB (5), initiating the expression of pro-inflammatory cytokines such as IL-1β and IL-6, and TNF-α (6). The production of pro-inflammatory cytokines and the action of CD14 stimulate the processing of antigens and their presentation to the MHC (7), which results in the maturation of APCs, as well as the activation of IFN-γ-producing CD4+ T lymphocytes (8), which leads to polarization towards Th1 (9).
Vaccines 14 00054 g005
Figure 6. CpG ODN is phagocytosed by B cells and pDCs (1), processed and recognized in the endosome by TLR-9 (2). TLR-9 stimulation leads to a cascade of MyD88, IRAK, and TRAF-6 signaling (3), which activates NF-κB (4), producing proinflammatory cytokines (5), which lead to the maturation of naïve lymphocytes to the Th1 profile (6), and the secretion of IFN-γ (7). IFN-γ production by CD4+ Th1 lymphocytes leads to the activation of antigen-specific CD8+ T cells (8). The pDCs express TNF-α, IFN-ꞵ, and IFN-α, activating and recruiting NK, monocytes, and neutrophils (9). Activated B cells increase the production of IL-6 and IL-12, and secrete the immunoglobulins IgM and IgG (10).
Figure 6. CpG ODN is phagocytosed by B cells and pDCs (1), processed and recognized in the endosome by TLR-9 (2). TLR-9 stimulation leads to a cascade of MyD88, IRAK, and TRAF-6 signaling (3), which activates NF-κB (4), producing proinflammatory cytokines (5), which lead to the maturation of naïve lymphocytes to the Th1 profile (6), and the secretion of IFN-γ (7). IFN-γ production by CD4+ Th1 lymphocytes leads to the activation of antigen-specific CD8+ T cells (8). The pDCs express TNF-α, IFN-ꞵ, and IFN-α, activating and recruiting NK, monocytes, and neutrophils (9). Activated B cells increase the production of IL-6 and IL-12, and secrete the immunoglobulins IgM and IgG (10).
Vaccines 14 00054 g006
Figure 7. Immunogenicity of the S20 ribosomal antigen formulated as a DNA vaccine. BALB/c mice were immunized with the third-generation vaccine produced with the S20 ribosomal gene inserted in the pVAX-1 plasmid. BALB/c mice were immunized three times with 100 mg/animal/animal intramuscularly. Fifteen days after the last injection, the T cell and macrophage responses were analyzed. In this case, pVAX1-S20 reduced the Th1 and CD8+ T activities, allowing the multiplication of amastigote forms in macrophages that produced low amounts of IL-12 cytokines.
Figure 7. Immunogenicity of the S20 ribosomal antigen formulated as a DNA vaccine. BALB/c mice were immunized with the third-generation vaccine produced with the S20 ribosomal gene inserted in the pVAX-1 plasmid. BALB/c mice were immunized three times with 100 mg/animal/animal intramuscularly. Fifteen days after the last injection, the T cell and macrophage responses were analyzed. In this case, pVAX1-S20 reduced the Th1 and CD8+ T activities, allowing the multiplication of amastigote forms in macrophages that produced low amounts of IL-12 cytokines.
Vaccines 14 00054 g007
Table 1. Summary of experimental models used in visceral leishmaniasis.
Table 1. Summary of experimental models used in visceral leishmaniasis.
Animal ModelCausative AgentKey Features of VL InfectionRelevance to Human VLLimitationsReferences
Dog (Canis familiaris)L. (L.) infantumNatural domestic reservoir
Infection mimics human VL clinical progression
Symptoms: weight loss, anemia, lymphadenopathy, fever
Skin lesions present (absent in humans)
Parasites in spleen and skin even in asymptomatic dogs
Similar disease progression
Important for epidemiology and experimental models
Useful for vaccine and drug testing
High maintenance cost
Ethical concerns
Limited animal numbers per study
[23,24,25,26,27,30,35]
Mouse (mus musculus)L. (L.) donovani and L. (L.) infantumSubclinical and limited infection
Effective cellular immune response
Chronic fever, hepatosplenomegaly, cachexia absent
Useful for studying protective immunity and genetic factors
Model for self-controlled or oligosymptomatic human cases
Widely used due to availability of immunological tools
Does not fully reproduce human VL pathology[36,37]
Golden hamster (mesocricetus auratus)L. (L.) donovaniHighly susceptible to Leishmania
Develops cachexia, hepatosplenomegaly, pancytopenia, hypergammaglobulinemia
Granulomas in liver, progressive spleen infection
Failure to produce NO despite Th1 cytokine response
- Ascites and renal complications rare in humans
Reproduces many human VL clinical and immunopathological featuresLimited immunological reagents
Some pathological features not typical of humans
[28,29,38,39]
Rhesus macaque (Macaca mulatta)L. (L.) infantumSystemic disease similar to human VL
Symptoms: fever, diarrhea, weight loss, anemia, hypergammaglobulinemia, lymphadenopathy, hepatosplenomegaly
Hepatic granuloma formation with epithelioid and Langhans giant cells
Useful for vaccine and drug evaluation
High phylogenetic similarity
Similar immune responses
Mimics human infection
High cost and ethical issues
Complex maintenance
Some differences in disease control
[34]
Cebus apella monkeyL. (L.) infantum chagasiTransient infection
Effective cellular immune response controls parasite spread
Not recommended to mimic natural infection but useful to study resistance mechanismsDoes not adequately mimic human natural infection[40]
VL—visceral leishmaniasis.
Table 2. Summary of experimental models used in cutaneous leishmaniasis.
Table 2. Summary of experimental models used in cutaneous leishmaniasis.
Experimental ModelCausative AgentHuman Clinical Form(S)Model CharacteristicsRelevance to Human DiseaseLimitationsReferences
MOUSE (MUS MUSCULUS, BALB/C)L. (L.) amazonensisLCL
MCL (rare)
DCL
Progressive ulcerative lesions cutaneous metastasis
Th2-type immune response associated with susceptibility
Mimics progressive disease in some human cases
Useful for immunological studies
Limited chronic disease features[46]
MOUSE (MUS MUSCULUS, C57BL/10, C57BL/6, DBA/2)Chronic disease or subclinical infection depending on strain
Eosinophil and mast cell infiltration.
Th1 or Th2 immune responses
Mimics spectrum of human disease
Allows study of genetic and immune response variability
Strain-dependent variability; some immune responses differ from humans[47,48]
RHESUS MONKEYS (MACACA MULATTA) Progressive ulcerative lesions
Lesions display mononuclear cell infiltrates composed of macrophages, lymphocytes, and plasma cells, along with tuberculoid-type granuloma formation
There is a shift in circulating T cell subpopulations, initially a predominance of CD4+ T cells, followed by an increase in CD8+ T cells as the infection progresses
Mimics progressive of the cutaneous leishmaniasis in humans
Useful for immunological studies
Useful to evaluate the efficacy of new vaccines
High cost and ethical issues
Complex maintenance
Limited availability and sample sizes
Longer experimental timelines
[49]
MOUSE (MUS MUSCULUS, BALB/C) L. (V.) braziliensis LCL
MCL
Transient or ulcerated lesions depending on inoculation site (hind footpad or ear) Some infection features resemble human disease including lesion ulceration and parasite persistence Resistance in many mouse strains; difficulty obtaining infectious stages [42,51]
SAPAJUS APELLA MONKEYS Parasite persistence during infection
Cutaneous lesions present
Mimics human infection dynamics well.
Useful for immunopathology and vaccine studies
High cost and ethical issues
Complex maintenance
Limited availability and sample sizes
Longer experimental timelines
[50]
MOUSE (MUS MUSCULUS, BALB/C) L. (V.) panamensis LCL
Rare mucosal involvement
Lower virulence observed in rodents Limited data
Less commonly used models
Low virulence limits study of full disease spectrum [43]
MOUSE (MUS MUSCULUS, BALB/C) L. (V.) shawi LCL Higher susceptibility with footpad swelling
Severe inflammation
Th1 response
Represents natural infection in humans
Useful for drug and vaccine development
Species-specific response; may not represent all human cases [12]
LCL: Localized cutaneous leishmaniasis; MCL: Mucocutaneous leishmaniasis; DCL: Diffuse cutaneous leishmaniasis.
Table 3. Leishmanization against leishmaniasis over the past 20 years.
Table 3. Leishmanization against leishmaniasis over the past 20 years.
NameVaccine ParasiteAdjuvantHostRouteDosesChallengeProtectionReference
LdL. donovani BALB/cSubcutaneousSingle doseL. donovani<50% less liver parasite burden[120]
LiL. infantum DogSubcutaneousSingle doseL. infantum100% without Leishmania DNA; 97.8% without clinical signs[115]
Li HSP70-II−/−L. infantum BALB/cSubcutaneousSingle doseL. major90% less spleen parasite burden[117]
LmL. majorCpGC57BL/6IntradermalSingle dose 100% without lesion[110]
LmL. major BALB/cSubcutaneousSingle doseL. major50% less skin parasite burden[122]
Lm Cen−/−L. major HamsterIntradermalSingle doseL. donovani90% less liver and spleen parasite burden[118]
Lm tkcd+/+L. major C57BL/6SubcutaneousSingle doseL. major90% spleen parasite burden; 100% without lesion[116]
LtL. tarentolae BALB/cIntraperitonealSingle doseL. donovani80% spleen and liver parasite burden[113]
LtL. tarentolaeCpGBALB/cIntraperitonealSingle doseL. major87% spleen parasite burden[114]
LtL. tarentolae BALB/cIntraperitonealSingle doseL. infantum90% spleen and liver parasite burden[112]
Table 4. First-generation vaccines against leishmaniasis over the past 20 years.
Table 4. First-generation vaccines against leishmaniasis over the past 20 years.
NameVaccine ParasiteAdjuvantHostRouteDosesChallengeProtectionReference
KBMA LiL. infantum BALB/cSubcutaneous3 dosesL. infantum30% less liver parasite burden[138]
LaL. amazonensis C57BL/6Intranasal2 dosesL. amazonensis50% less lesion size and parasite burden[139]
LaL. amazonensisADDAVAX®C57BL/6Intranasal2 dosesL. amazonensisIneffective[139]
LaL. amazonensisSaponinBALB/cSubcutaneous3 dosesL. chagasi25% less liver parasite burden; 50% less spleen parasite burden[128]
La-phVac and La-mtVacL. amazonensis C57BL/10Subcutaneous2 dosesL. amazonensis50% without lesion[123]
LbL. braziliensisSaponinDogSubcutaneous3 doses Indeterminate[129]
LbL. braziliensisSaponinBALB/cSubcutaneous3 dosesL. chagasi25% less liver parasite burden; 50% less spleen parasite burden[128]
Lb-SalL. braziliensisSaponinDogSubcutaneous3 doses Indeterminate[130]
LcL. chagasiSaponinBALB/cSubcutaneous3 dosesL. chagasi90% less liver and spleen parasite burden[140]
LdL. donovani BALB/cIntraperitoneal3 dosesL. donovani15% less liver parasite burden; 24% less spleen parasite burden[141]
LdL. donovaniBCGBALB/cIntraperitoneal3 dosesL. donovani30% less liver parasite burden; 40% less spleen parasite burden[141]
LdL. donovaniMPL/TDMBALB/cIntraperitoneal3 dosesL. donovani30% less liver parasite burden; 40% less spleen parasite burden[141]
Ld-LipL. donovaniCationic liposomes Intraperitoneal3 dosesL. donovani80% less liver parasite burden; 60% less spleen parasite burden[141]
Leishvacin®L. amazonensis HumanIntramuscular3 dosesL. panamensisIndeterminate[132]
LiL. infantumBCGDogSubcutaneous3 doses Indeterminate[142]
LiL. infantumAdjuPrimeTMDogSubcutaneous3 doses Indeterminate[142]
LiL. infantumMPL/TDM/CWSDogSubcutaneous3 doses Indeterminate[142]
LmL.majorBCGBALB/cSubcutaneous3 dosesL. major50% less lesion size; 90% less skin parasite burden[124]
LmL. majorAlumBALB/cSubcutaneous3 dosesL. major50% less lesion size; 70% less skin parasite burden[124]
LmL.majorMontanide ISA 720BALB/cSubcutaneous3 dosesL. major80% less lesion size; 90% less skin parasite burden[124]
LmL.majorBCGHumanIntradermalSingle dose Indeterminate[137]
LmL. majorCpGBALB/cSubcutaneous3 dosesL. major50% less skin parasite burden[122]
Lm(FKP)L. major BALB/cSubcutaneous3 dosesL. major50% less lesion size; 30% less skin parasite burden[124]
sTLAVacL. amazonensis C57BL/6Subcutaneous2 dosesL. amazonensisIndeterminate[125]
LsAmaAgL. shawi BALB/cSubcutaneous2 dosesL. shawiIneffective[134]
LsProAgL. shawi BALB/cSubcutaneous2 dosesL. shawi60% less lesion size; 70% less skin parasite burden[134]
WPVL. amazonensis and L. braziliensisBCGDogSubcutaneous3 doses Indeterminate[131]
Table 5. Second-generation vaccines against leishmaniasis over the past 20 years.
Table 5. Second-generation vaccines against leishmaniasis over the past 20 years.
NameVaccine ParasiteAdjuvantHostRouteDosesChallengeProtectionReference
LsPass1L. shawi BALB/cSubcutaneous2 dosesL. shawiIneffective[157]
LsPass2L. shawi BALB/cSubcutaneous2 dosesL. shawi50% less lesion size; 70% less skin parasite burden[157]
LsPass3L. shawi BALB/cSubcutaneous2 dosesL. shawi75% less lesion size; 90% less skin parasite burden[157]
LiRic1L. infantum BALB/cIntraperitoneal4 dosesL. infantum50% less spleen parasite burden[156]
LiRic2L. infantum BALB/cIntraperitoneal4 dosesL. infantum66% less spleen parasite burden[156]
CaniLeish®/ESPL. infantumQA-21DogSubcutaneous3 doses 92.7% healthy[161]
ChimeraTLeishmaniaSaponinBALB/cSubcutaneous3 dosesL. infantum80% less liver and spleen parasite burden; 90% less bone marrow and lymph node parasite burden[155]
His6/LACKL. braziliensisCpG+AlumenBALB/cIntraperitoneal3 dosesL. braziliensisIneffective[177]
His6/LbSTIL. braziliensisCpG+AlumenBALB/cIntraperitoneal3 dosesL. braziliensisIneffective[177]
His6/LeIFL. braziliensisCpG+AlumenBALB/cIntraperitoneal3 dosesL. braziliensisIneffective[177]
His6/TSA,L. braziliensisCpG+AlumenBALB/cIntraperitoneal3 dosesL. braziliensisIneffective[177]
LaPSA-38SL. amazonensis In vitro Indeterminate[178]
LaSPL. amazonensisSaponinBALB/cIntramuscular3 dosesL. amazonensisIneffective[179]
Ld31L. donovaniCationic liposomesBALB/cIntraperitoneal3 dosesL. donovani70% less liver and spleen parasite burden[152]
Ld51L. donovaniCationic liposomesBALB/cIntraperitoneal3 dosesL. donovani70% less liver and spleen parasite burden[152]
Ld72L. donovaniCationic liposomesBALB/cIntraperitoneal3 dosesL. donovani60% less liver and spleen parasite burden[152]
Ld78L. donovaniCationic liposomesBALB/c 3 dosesL. donovani97% less liver parasite burden[152]
Ld79L. donovaniMPLBALB/c 3 dosesL. donovani96% less liver parasite burden[153]
Ld91L. donovaniCationic liposomesBALB/cIntraperitoneal3 dosesL. donovani40% less liver and spleen parasite burden[152]
LdGP63L. donovaniCationic liposomesBALB/cIntraperitoneal3 dosesL. donovani80% less liver and spleen parasite burden[147]
LdPDIL. donovani HamsterIntramuscular3 dosesL. donovani90% less spleen, liver and bone marrow parasite burden[180]
LdrA2L. donovaniSaponinDogSubcutaneous3 doses 43% no parasite burden detected; 72% asymptomatic[178]
LdTPIL. donovani HamsterIntramuscular3 dosesL. donovani~90% less spleen and liver parasite burden[181]
Leish-111f(MML) or LEISH-F1LeishmaniaCpGC57BL/6Subcutaneous3 dosesL. major83% less lesion size; 45% less skin parasite burden[158]
Leish-111f(MML) or LEISH-F1LeishmaniaMPL-SEDogIntradermal3 dosesL. infantum29% healthy[146]
Leish-111f(MML) or LEISH-F1LeishmaniaMPL-SEDogSubcutaneous3 dosesL. infantumIneffective[90]
Leish-111f(MML) or LEISH-F1LeishmaniaAdjuprimeDogSubcutaneous3 dosesL. infantumIneffective[90]
Leish-111f(MML) or LEISH-F1LeishmaniaMPL-SEHumanSubcutaneous3 doses Indeterminate[182]
Leish-111f(MML) or LEISH-F1LeishmaniaMPL-SEHumanSubcutaneous3 doses Indeterminate[183]
Leishmune®L. donovani DogSubcutaneous3 doses 100% asymptomatic[184]
Leishmune®/FMLL. donovaniSaponinDogSubcutaneous3 doses 89% healthy[166]
Leishmune®/FMLL. donovaniSaponinDogSubcutaneous3 doses 100% asymptomatic; 100% without Leishmania DNA[162]
Leishmune®/FMLL. donovaniSaponinDogSubcutaneous3 doses Indeterminate[165]
LeishTec®/rA2LeishmaniaSaponinDogSubcutaneous3 doses 92% healthy[166]
LeishTec®/rA2LeishmaniaSaponinDogSubcutaneous3 doses 69% healthy[160]
LeishTec®/rA2LeishmaniaSaponinDogSubcutaneous3 doses 63.7% healthy[167]
LetiFend®/Q proteinL. infantum DogSubcutaneousSingle dose 95.3% healthy[159]
Q proteinLeishmania DogSubcutaneousSingle dose 57% asymptomatic[172]
Q proteinLeishmania DogSubcutaneous2 doses 28% asymptomatic[172]
Li ESAp-MDPL. infantumMDPDogSubcutaneous2 dosesL. infantum100% less bone marrow parasite burden[176]
Li ESAp-MDPL. infantumMDPDogSubcutaneous2 dosesL. infantum100% less bone marrow parasite burden[174]
Li ESAp-MDPL. infantumMDPDogSubcutaneous2 doses 100% less bone marrow parasite burden; 91% without Leishmania DNA[175]
LiH1L. infantumMontanide ISA 720DogIntradermal3 dosesL. infantum62,5% healthy[146]
LiH1-HASPB1L. infantumMontanide ISA 720DogIntradermal3 dosesL. infantum50% healthy[146]
LiH1-SMT-Hy-SAPL. infantum BALB/cSubcutaneous2 dosesL. infantum80% less spleen parasite burden[154]
LiH1-SMT-Hy-SAPL. infantumFreundBALB/cSubcutaneous2 dosesL. infantum80% less spleen parasite burden[154]
LiHASPB1L. infantumMontanide ISA 720DogIntradermal3 dosesL. infantum50% healthy[146]
LiPHBL. infantumSaponinBALB/cSubcutaneous3 dosesL. infantum60% less liver, spleen and lymph node parasite burden; 75% less bone marrow parasite burden[185]
LmLACKL. major BALB/cSubcutaneous2 dosesL. majorIneffective[144]
LmTRYPL. major BALB/cSubcutaneous2 dosesL. major20% less lesion size[144]
LPG3-IFAL. chagasiSaponinBALB/cSubcutaneous3 dosesL. chagasi96% less liver and spleen parasite burden[186]
LPG3-SapL. chagasiSaponinBALB/cSubcutaneous3 dosesL. chagasi98% less liver and spleen parasite burden[186]
NGP5B/αGal CpGC57BL/6Subcutaneous4 dosesL. major72% less skin parasite burden[187]
rPHBL. infantumSaponinBALB/cSubcutaneous3 dosesL. infantum60% less liver parasite burden; 66% less spleen parasite burden and lymph node; 75% less parasites in the bone marrow;[185]
LdGP63L. donovani BALB/cSubcutaneous2 dosesL. donovani85% less spleen parasite burden; 68% less liver parasite burden[150]
LdHsp70L. donovani BALB/cSubcutaneous2 dosesL. donovani50% less spleen parasite burden; 40% less liver parasite burden[150]
LdGP63-Hsp70L. donovani BALB/cSubcutaneous2 dosesL. donovani90% less spleen parasite burden; 94% less liver parasite burden[150]
LdGP63-Hsp70L. donovaniMPL-ABALB/cSubcutaneous2 dosesL. donovani94% less spleen parasite burden; 97% less liver parasite burden[151]
LdGP63-Hsp70L. donovaniALDBALB/cSubcutaneous2 dosesL. donovani92% less spleen parasite burden; 93% less liver parasite burden[151]
LdLACKL. donovani BALB/cIntraperitoneal3 dosesL. donovaniIneffective[145]
LdLACKL. donovaniLiposomeBALB/cIntraperitoneal3 dosesL. donovaniIneffective[145]
LdGP63L. donovaniLiposomeBALB/cIntraperitoneal3 dosesL. donovani66% less spleen parasite burden; 71% less liver parasite burden[145]
Table 6. Third-generation vaccines against leishmaniasis over the past 20 years.
Table 6. Third-generation vaccines against leishmaniasis over the past 20 years.
Vaccine NameParasite SpeciesAdjuvantHostRouteDosesChallengeProtectionReference
pCMV/gp63L. major-BALB/cIntramuscularSingle doseL. major100× less parasites tan control[193]
pcDNA3.1-gp63L. donovaniCpGBALB/cIntramuscular2 doses (DNA vaccine)
2 doses (recombinant protein)
L. donovani
L. major
~105–6-fold reduction in the spleen and 103 in the liver
Lesion size reduction ~2.5
[195]
pcDNA3.1-gp63L. mexicana-BALB/cGene gun immunization2 dosesL. mexicana88% less lesion size[197]
gp63 amino acid sequence (138–360)L. infantum-BALB/cIntramuscular and subcutaneous2 dosesL. infantum88% less spleen parasite burden[199]
pcDNA3-LACKL. majorrIL-12BALB/cSubcutaneous2 dosesL. major~50% less lesion size; 18% less lymph node parasite burden[200]
pcDNA3.1-LACKL. donovani-BALB/cIntradermal2 dosesL. donovaniIneffective[202]
pCI-neo-LACKL. chagasi-BALB/cIntramuscular2 dosesL. chagasiIneffective[203]
pCI-neo-LACKL. amazonensis-BALB/cIntranasal2 dosesL. amazonensis66% less lesion size; 83% less skin parasite burden[204]
pCI-neo-LACKL. infantum-BALB/cIntranasal2 dosesL. infantum90% less liver parasite burden; 85% less spleen parasite burden[205]
pCI-neo-LACKL. infantum-HamsterIntranasal2 dosesL. infantum88% less spleen parasite burden; 75 less liver parasite burden.[206]
pCI-neo-LACKL. amazonensisCrosslinked chitosan microparticlesBALB/cIntranasal2 dosesL. amazonensis51% less lesion size; 99% less skin parasite burden[208]
MVA-LACKL. major-BALB/cIntradermal and Intraperitoneal2 dosesL. major90% less lesion size; 80% less lymph node parasite burden[209]
MVA-LACKL. infantum-HamsterIntramuscular2 dosesL. infantum66% less liver parasite burden; 83% less bone marrow parasite burden;[206]
pORT-LACKL. infantum-DogsSubcutaneous2 dosesL. infantum25% negative for real-time PCR; 62.5% with only one disease sign[210]
pPAL-LACKL. infantum-DogsIntranasal2 dosesL. infantum60% without disease sign; 92% less bone marrow parasite burden; 80% less spleen parasite burden; 42% less liver parasite burden[211]
pcDNA3-LiP0L. infantum-BALB/cIntramuscular2 dosesL. major44% less lesion size; 84% less lymph node parasite burden.[215]
pcDNA3-LiP0L. infantum-HamsterIntramuscular3 dosesL. infantum99% less lymph node parasite burden; 99.9% less spleen parasite burden[216]
pVAX1-P1L. donovani-HamsterIntramuscular2 dosesL. donovani75.69% less spleen and liver parasite burden[72]
pVAX1-S20--BALB/cIntramuscular3 doses-IneffectiveData not published
pcDNA3-TRL. major-BALB/cSubcutaneous3 dosesL. major22% less lesion size[218]
MVA-TRL. major-BALB/cSubcutaneous3 dosesL. major22% less lesion size[218]
pVAX-1-TR + MVAL. (V.) panamensis BALB/cIntradermal and Intraperitoneal3 dosesL. (V.) panamensis77% les skin parasite burden; 90% less lymph node parasite burden[219]
pcDNA-SODL. donovaniCpG ODNBALB/cIntramuscular2 dosesL. major33% less lesion size[224]
pcDNA-GMCSF-SODL. donovaniCpG ODNBALB/cIntramuscular2 dosesL. major22% less lesion size[224]
pVAX1-SODL. amazonensis-BALB/cIntramuscular3 dosesL. amazonensis42% less lesion size; 71% less skin parasite burden[225]
pcDNA3-TSAL. major-BALB/cIntramuscular3 dosesL. major71% less lesion size; 75% less skin parasite burden[226]
pcDNA3-TSA-LmSTIL. major-BALB/cIntramuscular3 dosesL. major80% less lesion size[228]
Leishmania multicomponent (10 antigen)L. donovani-DogsIntramuscular3 doses--[229]
LEISHDNAVAXLeishmania-BALB/cIntradermal3 dosesL. donovani94% less liver parasite burden; 91% less spleen parasite burden[230]
pMOK-Kmp11/-TRYP/-LACK/-GP63L. infantum-DogsIntradermal4 dosesL. infantumIneffective[231]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Passero, L.F.D.; Cavallone, I.N.; Araújo Flores, G.V.; de Lima Melchert, S.S.; Laurenti, M.D. An Overview of the Factors Related to Leishmania Vaccine Development. Vaccines 2026, 14, 54. https://doi.org/10.3390/vaccines14010054

AMA Style

Passero LFD, Cavallone IN, Araújo Flores GV, de Lima Melchert SS, Laurenti MD. An Overview of the Factors Related to Leishmania Vaccine Development. Vaccines. 2026; 14(1):54. https://doi.org/10.3390/vaccines14010054

Chicago/Turabian Style

Passero, Luiz Felipe Domingues, Italo Novais Cavallone, Gabriela Venicia Araújo Flores, Sarah Santos de Lima Melchert, and Márcia Dalastra Laurenti. 2026. "An Overview of the Factors Related to Leishmania Vaccine Development" Vaccines 14, no. 1: 54. https://doi.org/10.3390/vaccines14010054

APA Style

Passero, L. F. D., Cavallone, I. N., Araújo Flores, G. V., de Lima Melchert, S. S., & Laurenti, M. D. (2026). An Overview of the Factors Related to Leishmania Vaccine Development. Vaccines, 14(1), 54. https://doi.org/10.3390/vaccines14010054

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop