4,746
Views
13
CrossRef citations to date
0
Altmetric
Review

From infection to vaccination: reviewing the global burden, history of vaccine development, and recurring challenges in global leishmaniasis protection

, , , , , , & show all
Pages 1431-1446 | Received 04 Jul 2021, Accepted 13 Aug 2021, Published online: 15 Sep 2021

ABSTRACT

Introduction

Leishmaniasis is a major public health problem and the second most lethal parasitic disease in the world due to the lack of effective treatments and vaccines. Even when not lethal, leishmaniasis significantly affects individuals and communities through life-long disabilities, psycho-sociological trauma, poverty, and gender disparity in treatment.

Areas covered

This review discusses the most relevant and recent research available on Pubmed and GoogleScholar highlighting leishmaniasis’ global impact, pathogenesis, treatment options, and lack of effective control strategies. An effective vaccine is necessary to prevent morbidity and mortality, lower health care costs, and reduce the economic burden of leishmaniasis for endemic low- and middle-income countries. Since there are several forms of leishmaniasis, a pan-Leishmania vaccine without geographical restrictions is needed. This review also focuses on recent advances and common challenges in developing prophylactic strategies against leishmaniasis.

Expert opinion

Despite advances in pre-clinical vaccine research, approval of a human leishmaniasis vaccine still faces major challenges – including manufacturing of candidate vaccines under Good Manufacturing Practices, developing well-designed clinical trials suitable in endemic countries, and defined correlates of protection. In addition, there is a need to explore Challenge Human Infection Model to avoid large trials because of fluctuating incidence and prevalence of leishmanasis.

1. Introduction

Due to the increased recognition of neglected tropical diseases (NTDs) as major public health problems, great strides have been made toward reducing the burden of these infectious diseases in developing countries around the world. However, leishmaniasis remains the second most lethal parasitic disease due to the substantial obstacles to universal treatment in many endemic areas and the lack of an approved vaccine [Citation1].

2. Methods

Pubmed and GoogleScholar databases were searched to compile the most relevant and up-to-date information on global leishmaniasis etiology, burden, prevention and treatment alternatives to vaccination, historical vaccine candidates, and previous vaccine candidate performance. Due to the scope of the article, not all significant studies in Leishmania literature could be cited.

3. Leishmaniasis, a global public health problem

3.1. Pathogenesis of Leishmaniasis

Leishmaniasis encompasses multiple disease presentations in humans caused by unicellular, motile protozoan parasites of the genus Leishmania. Leishmania species have two major life stages – motile promastigotes and immotile amastigotes. Promastigotes reside in sandflies and are spread by at least 98 sandfly species (from the subfamily Phlebotominae) which serve as intermediate host and vector [Citation2], whereas amastigotes are responsible for disease symptomology in infected people via intracellular replication, manipulations of the host immune system, and secondary infections [Citation3].

Although the fundamentals of the dimorphic lifecycle of Leishmania in host and sandfly is the same for all forms of leishmaniases, disease pathology differs by Leishmania species and geographical location of infection. Clinical presentation of leishmaniasis is classified into three main types: cutaneous leishmaniasis (CL), visceral leishmaniasis (VL; also known as Kala-azar), and mucocutaneous leishmaniasis (MCL) [Citation4]. VL is the most serious public health concern among the leishmaniases as it is highly lethal if left untreated due to its systemic effects. Initial symptoms include fever, hepatosplenomegaly, and low levels of erythrocytes and leukocytes as well as platelets. However, the most deadly effects include later sepsis development from secondary infection, excessive bleeding from loss of platelets, severe anemia, and rapid weight loss [Citation5]. Depending on geographic distribution of species, VL can be caused by L. donovani or L. infantum – the latter of which can also infect dogs in Europe, Brazil, and North Africa [Citation5]. Along with dogs, other animals can develop infections from multiple human Leishmania spp. including cats, horses, sheep, hyrax, snakes, lizards, sloths, foxes, and numerous rodents such as the fat sand rat [Citation6–15]. Furthermore, with some regional differences, roughly 10% of all dogs in Southwestern Europe and 2–50% of dogs in various regions of Brazil are estimated to be carriers of canine leishmaniasis, therefore public health strategies to control human leishmaniasis must and do consider the possibility of zoonotic reservoir hosts for Leishmania species [Citation16–18].

Unlike VL, MCL and CL are not lethal but still cause severe disease which can result in social stigma for patients due to serious disfigurement and permanent scars. Skin malformities in CL patients are diverse – often simulating herpes zoster or leprosy lesions, among other potential misdiagnoses [Citation19]. Although this diversity in clinical manifestations can make diagnosis exceedingly difficult, symptoms typically begin with a small papule at the site of infection which then grows into a larger skin nodule and subsequently to an open ulcer [Citation19]. Depending on the causative species of CL, lesions can be localized, diffuse, or disseminated, and can be self-healing in 3–24 months or become chronic in <10% of patients (as in leishmaniasis recidivans) [Citation20]. Each distinct CL lesion has a unique immune response profile [Citation21]. Another distinct form of leishmaniasis is MCL. Similarly, to CL, MCL causes destructive changes to skin around mucous membranes of the host, especially the nose and mouth [Citation22].

3.2. Diagnosis and treatment of Leishmaniasis

Although preventive measures through government and individual action can be effective, they require consistent investment to reduce disease risk for local populations, therefore rapid diagnosis is equally important to high-endemic regions. The gold-standard for VL and CL diagnosis remains microscopy of biopsy or aspirate samples in the case of VL or histology/direct microscopy in the case of CL [Citation23,Citation24]. Other tests are becoming more common such as rapid diagnostic antibody/antigen tests, latex agglutination, and PCR which are further discussed here [Citation25,Citation26]. These alternatives are especially appealing to remote medical centers seeking decentralized point-of-care options, however antibody tests in particular are limited in their utility for relapsed infections [Citation27]. Furthermore, PCR is increasingly important to identify specific species of infection for tailored treatment of patients [Citation28].

After diagnosis, treatment of leishmaniasis must also contend with multiple challenges. Unfortunately, although affordable, the WHO-standard pentavalent antimonial combination regimen can cause severe side effects such as liver, heart, and kidney toxicity [Citation1,Citation22,Citation27,Citation29,Citation30]. In addition, drug efficacy varies by region due to parasite resistance to first line antimonials which has been observed as high as 40–60% in Algeria and Bihar, India [Citation31,Citation32]. Intravenous application of liposomal amphotericin B represents the most significant advance from this paradigm in recent years as it can clear all systemic leishmaniases with low levels of toxicity – however, at much greater expense [Citation27,Citation29,Citation33]. Additional treatment alternatives have been under investigation including surgery [Citation34], cryosurgery [Citation35,Citation36], heat therapy [Citation37–39], as well as a variety of localized drug-delivery strategies for localized lesions [Citation22]. Lastly, plastic surgery can attempt to rectify disfiguring scars that often remain following clearance of CL and MCL [Citation40].

3.3. Immunology of Leishmaniasis

Given the array of parasite species which cause leishmaniases around the globe and diverse forms of presentation, the immunology of these diseases is complex. As such, here we review only the `main concepts of immunology for the major forms of leishmaniases while providing more in-depth resources on each species here [Citation21,Citation41–47].

Leishmania parasites can be acquired by a broad range of phagocytic cell populations including neutrophils, dendritic cell, and monocytes; however, Leishmania’s preferred host cell is the macrophage. Regardless of locality, these cells phagocytize the promastigotes deposited by the sandfly and facilitate parasite differentiation into amastigotes as well as parasite replication within the host cell phagolysosome. Leishmania parasites prevent the infected cell from undergoing apoptotic clearance to allow the host macrophage to assist the amastigote’s spread into new cells via membranous extrusions, which attract and are internalized by nearby uninfected macrophages [Citation48]. This colonization of macrophages occurs most heavily in bone marrow, spleen, liver, and lymph nodes for VL, primarily in the skin for CL, or mucosa for MCL [Citation19,Citation41]. In all leishmaniases, heavily parasitized macrophages can finally become successful in killing amastigotes primarily following activation by CD4+ Th1 cells [Citation41,Citation49]. In response to MHC Class II-facilitated antigen presentation from the infected macrophage phagolysosomes, CD4+ Th1 cells release IFN-γ. This activates the infected M1 macrophages to produce reactive oxygen species, nitric oxide and increase phagolysosome fusions to overcome the parasite defense mechanisms in both mouse and human infections [Citation50,Citation51]. Th2 immune response mechanisms characterized by the cytokines IL-4, IL-13, and improved antibody production also play roles in leishmaniasis; Th2 activation is generally counterproductive to CL clearance [Citation52,Citation53], but IL-4 production may be required for clearance in L. donovani infection during VL [Citation54,Citation55]. Paradoxically, both Th1 and Th2 responses can have mixed results for hosts depending on the overall balance [Citation56]. For example, although TNF-α and IFN-γ are beneficial Th1 cytokines in CL, excessive Th1 cytokine production can result in a hyperinflammatory state which enables the CL lesions to spread metastatically to nearby tissues [Citation57].

Regardless of the exact benefit to host for clearance, antibodies are inevitably made during Leishmania infection; this allows for seropositivity to be used for studying past infections. PCR and ELISA evidence from hyperendemic Bihar, India, revealed surprisingly high rates (~20%) of healthy individuals with no history of VL tested positive for both tests [Citation58]. This result can only be explained by previously unreported asymptomatic VL infections.

3.4. Biological risk factors for Leishmaniasis

Although many people are resistant, patients with confounding factors are especially at risk for leishmaniasis. Risks for susceptibility include acute and chronic malnourishment, HIV co-infection, host genetics, or individuals with prescribed medication for other health issues; these all involve immunosuppression and can enable dormant Leishmania to reactivate as well as present new opportunities for acute cases [Citation59–62]. In the case of HIV coinfection, the mode of action is predictable – HIV-mediated loss of CD4+ Helper T cells make clearance of disease more difficult through the loss of a core pathway for macrophage activation [Citation63], however the relationship between immunosuppression and leishmaniasis disease progression is still not fully understood [Citation62,Citation64].

As with host susceptibility, Leishmania virulence is also increasingly recognized for its variability even within a single species. Naturally occurring Leishmania strains with enhanced virulence have been found to distinguish themselves not only via typical single nucleotide polymorphisms (SNP) or indel type mutations in virulence proteins, but also via significant copy number variation of important genes [Citation65–68]. Copy number variation is the more unique challenge to leishmaniasis treatment as Leishmania parasites are fully capable of possessing variable chromosome ploidy and they regularly exchange genetic information – even across species such as in the creation of hybrid promastigotes or amastigotes [Citation69–72].

3.5. Epidemiology of Leishmaniasis

Given the diversity of host and pathogen biological concerns, research into demographic and geographic risk factors for leishmaniasis becomes even more compelling to fully understand the scope of at-risk populations. The burden of leishmaniasis is especially concentrated among 6 countries (Bangladesh, Brazil, Ethiopia, India, South Sudan, and Sudan) which contain >90% of VL cases and 10 countries (Afghanistan, Algeria, Brazil, Colombia, Costa Rica, Ethiopia, the Islamic Republic of Iran, Peru, Sudan, and the Syrian Arab Republic) with >70% of CL cases [Citation73]. Detailed maps classified by clinical form, Leishmania species vectors, reservoirs and regions, as well as interactive maps can be consulted in the following references [Citation74–76]. Per World Health Organization (WHO) 2016 estimates, 616 million people and 431 million people live in these high-burden endemic countries and are therefore at risk of VL and CL respectively [Citation73]. Furthermore, despite much work on improved access to diagnosis and treatments, 1.3 million total leishmaniasis cases were reported in 2015–1 million CL, 300,000 VL, and 20–50,000 lethal VL cases [Citation1,Citation77].

Among this large, susceptible population, demographic and environmental risk factors for VL and CL are diverse and complicated around the world [Citation78]. For example, VL risk factors were characterized in Ethiopia as biased toward the elderly, people who sleep outdoors for their occupation such as herders, displaced populations, as well as involving a variety of environmental factors [Citation79]. Other such studies in Kenya and Argentina also found that risk factors for vector contact strongly correlate to CL disease risk such as people who live near or work in forests, reside in homes with cracked walls or permanent openings from windows and roofing, reside near waterways or ponds, do not perform any type of insect control at their home, or are younger in age (<15) [Citation80,Citation81].

In aggregate, Hotez et al. estimated leishmaniases to be responsible for over 2 million Disability-Adjusted Life Years (DALYs) lost from death and disfigurements – 9th among all infectious diseases in 2004 [Citation82]. Although the bulk of this burden is concentrated in impoverished communities of tropical countries, significant developed countries must also be concerned with leishmaniases. Southern Europe such as Italy and Spain have always been endemic, however, following expansion of native sandfly ranges, the US has now also reported autochthonous cases in Texas, Oklahoma, Florida, and Arizona [Citation83–87]. Alongside this increased attention and awareness, significant progress has been made in reducing the number of deadly VL cases around the world – especially in India, Nepal, and Bangladesh – yet VL and CL remain serious public health threats globally due to the lack of a protective vaccine [Citation88,Citation89].

4. Critical need for a Leishmania vaccine

4.1. Strategies for the management of Leishmaniasis

NTDs such as leishmaniasis exert a heavy socio-economic burden on endemic countries with an already precarious health system and financial status [Citation90,Citation91]. Even when it is not lethal, this disease can have long lasting effects on affected individuals and communities including life-long disability, psycho-sociological trauma, poverty, and enhancing gender disparities [Citation92]. On an individual level, these issues impact the health, quality of life, and the ability of a person to work [Citation92,Citation93]. However, they can also present a significant financial burden for the government, as the implementation of effective disease prevention and control strategies can be prohibitively expensive, especially for low- and middle-income countries (LMIC) [Citation92,Citation94]. Despite the significant impact that these diseases have on public health and the economy of endemic countries, their global impact is limited, and they are often ignored by the international community [Citation94].

Over the years, the World Health Organization (WHO) has worked with the international community, as well as local governments, to manage human leishmaniasis by developing roadmaps with clear yearly goals for control and elimination. Some of the strategies implemented by this organization include developing preventive chemotherapy, intensifying disease management, strengthening vector control, and improving water availability and sanitation [Citation95]. Recently, WHO put forward a new approach highlighting the Sustainable Development Goals for each NTD for the years 2021–2030 [Citation96,Citation97]. The proposed target goal for VL is to eliminate this disease as a public health problem (<1% case fatality rate due to primary disease) in 64 countries. For CL, 85% of the cases have to be detected and reported in 87 countries, and 95% of these cases need to receive treatment by 2030 [Citation97].

Leishmania-specific strategic plans tailored to different geographical areas have been developed with the goals of improving surveillance, outbreak preparedness and response, diagnosis, treatment, rehabilitation, nutritional support, training and resources for medical professionals, control of sand fly vector and reservoir hosts, as well as reducing transmission, morbidity, and mortality in humans and dogs [Citation98,Citation99]. Unfortunately, many of these target goals have not yet been achieved, despite the efforts of the international community.

4.2. Challenges in the management of Leishmaniasis

Even with strong public health policies set in place in endemic areas, there are additional external factors that can hinder the achievement of the target goals by promoting leishmaniasis emergence and dissemination. For instance, military unrest and unstable political conditions can contribute to the limited implementation of preventive and curative measures [Citation100,Citation101]. Berry, et al. have described leishmaniasis as a ‘disease of guerrilla warfare,’ and showed an increased risk for CL and VL in areas with civil turmoil, ethnic conflicts, foreign military interventions, and political terror [Citation101]. Furthermore, the upsurge of international travel in the recent decades has increased the risk of exposure to leishmaniasis and other NTDs. In North, Central, and South America, 27% of CL cases have been recorded in areas bordering different countries [Citation102]. Tourists and military personnel traveling to endemic areas are especially at risk [Citation103]. Surveillance strategies such as GeoSentinel (Global Surveillance Network of the International Society of Travel Medicine in partnership with the US CDC), have been developed to monitor travel-related infections such as leishmaniasis [Citation104].

Numerous studies have also identified climate change as a predictor of leishmaniasis emergence and geographical distribution [Citation105–109]. Purse, et al. created a model to predict and quantify the role of climate change and other factors in determining the distribution of CL and VL in the Americas. Their results showed that climatic factors can explain up to 80% of the past disease variance [Citation106]. Another study predicted the ecological risk of contracting leishmaniasis to expand from Central America toward the United States and Southern Canada due to climate change [Citation109]. A similar trend was found in Europe using a forecasting niche modeling approach to investigate changes in disease dissemination. This model predicted that the range of sandfly distribution will expand north from the Mediterranean region as climatic conditions change [Citation105].

Animal reservoirs are another factor affecting disease emergence and transmission. Dogs remain the main reservoir for many forms of leishmaniasis, although other animals such as carnivores, rodents, and cattle can also be affected [Citation110,Citation111]. The seroprevalence of Leishmania in dogs ranges from 3 to 50% depending on the endemic areas, although PCR-based diagnostic methods suggest this percentage might be even higher [Citation112]. Culling seropositive dogs is recommended in many endemic countries, as treatment could lead to the development of drug resistance in Leishmania. However, culling has not been shown to be particularly effective to reduce incidence of human leishmaniasis, and it presents economic and ethical concerns [Citation113,Citation114]. Furthermore, almost half of all infected dogs do not present clinical symptoms, while the other half presents a wide range of nonspecific symptoms, making surveillance and diagnosis very difficult [Citation115]. Developing strategies to prevent and control canine leishmaniasis is not only a veterinary issue, but also a public health goal, as dogs can facilitate transmission to humans in endemic as well as Leishmania-free areas [Citation116]. This has been a particular problem in the Mediterranean region and South America [Citation112]. Although no vaccine has yet been approved for human use several vaccines are available for canine leishmaniasis, such as CaniLeish® and LetiFend® in Europe, and Leishmune® (withdrawn in 2014) and Leish-Tec® in Brazil. Safety and efficacy of these vaccines are reviewed in this reference [Citation117]. These observations suggest that improving surveillance and treatment of the disease might not be enough to significantly slow down the spread of leishmaniasis, and that there is a critical need for a strong campaign of prevention in the future decades.

4.3. Developing prophylactic strategies for the prevention of Leishmaniasis

The preventive strategies for human leishmaniasis to date are restricted to limiting exposure to the vector and preventing bites by using bed nets and insect repellent, wearing clothes that cover usually exposed areas of the body, and avoiding the outdoors at dusk and dawn when sand flies are most active [Citation91]. The development of effective prophylactic vaccines is imperative to control leishmaniasis. It is known that patients who recover from leishmaniasis are protected against subsequent infection [Citation118]. Moreover, prophylactic vaccination with a low dose of L. major in non-exposed areas of the body (leishmanization) has been previously employed in endemic areas as it elicits protective immunity against reinfection [Citation119,Citation120]. Although this practice remains unfeasible due to safety and standardization issues, leishmanization suggests that a wide variety of Leishmania antigens, as well as a low level of parasitic persistence, might be needed to provide long lasting immunity and indicates the feasibility of a vaccine [Citation120].

A recent paper used a mathematical transmission model to predict the impact of different vaccine candidates on human VL transmission and incidence in the Indian subcontinent. Overall, development of protective immunity leads to the fastest reduction in VL incidence. Assuming that 100% of the population is vaccinated and only VL and PKDL patients act as reservoirs, an annual incidence of less than 1 VL case per 10,000 individuals (elimination target) can be achieved in a moderately endemic area with a vaccine efficacy of about 60% [Citation121].

An important factor to consider when developing an effective human vaccine are asymptomatic individuals. From a surveillance and diagnostic standpoint, asymptomatic infections are difficult to discover and identify. Several studies have identified asymptomatic individuals as potential carriers for different species of Leishmania [Citation122–124]; however, a recent study conducted in the Indian state of Bihar shows that asymptomatic individuals do not play a major role in maintenance and transmission of L. donovani [Citation125]. Nevertheless, asymptomatic individuals infected with other species of Leishmania might display a greater degree of transmissibility to the sand fly. More research must be conducted to determine the impact these individuals have on disease transmission. Assuming that asymptomatic individuals are the main reservoir for human VL, a vaccine with only 37% efficacy could reduce infectiousness, although an efficacy of 70% would be needed to reduce the development of symptoms. To reach the elimination target, a vaccine must reduce infectivity of asymptomatic individuals by 50% for 11 years, or reduce infectivity of all affected individuals (symptomatic and asymptomatic) by 50% for 4 years. Furthermore, a vaccine that can reduce the chance of developing symptoms by 50% can eliminate VL in 10 years if only symptomatic individuals are responsible for disease transmission, and in 19 years if asymptomatic individuals are the main reservoir [Citation121].

Taken all together these studies highlight the difficulty of controlling NTDs such as leishmaniasis. Even when diagnosed in time, leishmaniasis is difficult to treat, as the therapeutic options currently available are limited and often present toxic side effects and increased parasitic resistance [Citation100]. Interestingly, individuals who recover from leishmaniasis acquire lifelong immunity against subsequent infections, indicating that a vaccine is feasible [Citation118]. Research also shows that development of an effective vaccine would be more economical than maintaining long-term vector control, diagnostic, and treatment strategies. Bacon, et al. used a computer model to assess the economic value of a CL vaccine in endemic Latin American countries. They have determined that even a vaccine with a limited efficacy of 50% and a modest duration of protection of 5 years would be more cost-effective than the current available chemotherapies [Citation126]. Similar results were found in relation to a VL vaccine in the Indian state of Bihar [Citation127]. A vaccination campaign would additionally prevent individuals from developing long-lasting disabilities, reducing the economic burden by lowering health care costs and eliminating lost productivity – elements that are especially important for financially disadvantaged governments and workers. This highlights the need for a pan-Leishmania vaccine effective against all (or most) species of Leishmania without geographical restrictions. Vector-based vaccines could help achieve this goal [Citation128]. Over the years the scientific community has developed different vaccine candidates, which can be classified into: killed or live-attenuated parasites, recombinant proteins, and DNA vaccines and are discussed below [Citation129].

5. Leishmania experimental vaccines to date

5.1. Killed Leishmania vaccines

First generation vaccines utilize antigens derived from the whole parasite, either in a killed or live-attenuated form. Leishmania can be cultured easily in cell-free media, which allowed for the initial use of whole killed Leishmania parasites in Leishmanin skin tests (LST) as a method to diagnose human patients [Citation130]. This previous use of Leishmania, as well as its simple and low cost of production in developing countries, spurred Sales-Gomes to explore different formulations of polyvalent, whole-killed Leishmania as a first-generation vaccine against CL in Brazil as early as 1939. Pessoa and Curban followed in the 1940s, observing 80% efficacy in clinical trials for a polyvalent vaccine consisting of 18 Leishmania strains [Citation131]. Later, in the 1970s, investigation was continued by Mayrink and colleagues in Brazil with a killed vaccine composed of five different non-specified Brazilian Leishmania isolates. These early studies showed the killed vaccine to be safe and immunogenic after two intramuscular injections, observed by a positive LST in 78.4% of vaccinated volunteers within 3 months. However, serological tests displayed a lack of circulating antibodies, indicating the ability of the vaccine to induce a cell-mediated response over a protective humoral response [Citation131–134]. First-generation Leishmania vaccines similar to this are the only vaccine candidates that have progressed to Phase 3 clinical trial evaluation [Citation135].

In Venezuela, Convit and colleagues introduced the use of Bacillus Calmette Guerin (BCG) as an adjuvant to autoclaved L. amazonensis to treat CL [Citation136]. Later studies utilized BCG as an adjuvant with killed Leishmania vaccines in an attempt to increase the cell-mediated response. Clinical studies have been conducted in Ecuador on a trivalent vaccine of L. braziliensis, L. guyanensis, and L. amazonensis [Citation137,Citation138] and a trivalent L. amazonensis vaccine known as Leishvacin [Citation139]. In Colombia, studies of a single strain L. amazonensis vaccine produced by BIOBRAS in Brazil have been investigated [Citation135,Citation140]. In Iran and Sudan, a L. major killed vaccine produced by the Razi Institute has also been tested [Citation141–147]. These candidates exemplify the ability of a killed Leishmania vaccine adjuvanted with or without BCG to be a safe option – safety being an advantage of killed vaccines. Many of these studies, as well as others that have been summarized [Citation131,Citation148,Citation149], have exhibited an induction of cell-mediated responses in vitro (high IFN-γ level) and in vivo (positive LST). However, in all cases except for the study by Armijos, et al. [Citation137], protection against future infection was not associated with vaccination; therefore, the lack of protection has been a disadvantage of killed vaccine designs. Thus, first generation of killed Leishmania parasites remain an inadequate vaccine candidate against leishmaniasis.

5.2. Subunit and recombinant vaccines

Second generation vaccines focus on pathogen antigens including purified native protein fractions, synthetic or recombinant subunits, and recombinant viruses or bacteria possessing antigen DNA [Citation148]. In the case of Leishmania, conservation of antigens amongst the various isolates and species, low antigenic variation, and the availability of the complete Leishmania genome sequence has allowed for the use of Leishmania proteins as vaccine candidates [Citation150]. Suitable peptide antigens can be identified through peptide mapping in which the whole parasite proteome, known peptide libraries, and known immunogenic proteins are screened in silico or in vitro [Citation151,Citation152]. The use of animal models such as mice, hamsters, dogs, and non-human primates have further aided in the discovery of many parasite antigens that induce protective immunity in such models [Citation153].

The numerous subunit and recombinant Leishmania vaccine candidates have been extensively reviewed [Citation148,Citation149,Citation153,Citation154]. These candidates utilize notable Leishmania antigens including gp63, p36/LACK, A-2, FML, PSA-2/gp46/M-2, LCR1, ORFF, KMP11, LeIF, LmSTI1, TSA, HASPB1, protein Q, and cysteine protease B(CPB) and A (CPA) [Citation148]. Many of these adjuvanted second-generation vaccine candidates have induced protection against reinfection in cutaneous animal models of L. major, L. mexicana, L. amazonensis, and L. pifanoi, as well as in visceral animal models of L. donovani, L. infantum, L. chagasi, and are therefore more efficacious than their killed Leishmania predecessors. The Infectious Disease Research Institute (IDRI) has generated a recombinant vaccine candidate, LEISH-F1, which was the first to progress to phase I and phase II clinical trials where it displayed safety and immunogenicity against L. major and L. infantum. Similar candidates produced by IDRI, LIESH-F2 (against CL) and LEISH-F3 (against L. donovani and L. infantum), have also entered clinical trials with promising results [Citation155–157]. Since pure proteins generally induce a weak cell-mediated response, adjuvants and multiple doses are often needed for these candidates to initiate and tailor long lasting vaccine-induced immunity – a disadvantage to subunit vaccines. In particular, VL patients who show an immunosuppressed state can be benefited using adjuvants. Aluminum salts seem to increase immunogenicity in murine models and in patients treated with sodium stibogluconate and have been proposed since the 1920s [Citation158]. Also, some liposomal mediators can be used to deliver antigens to antigen-presenting cells (APCs) [Citation159]. More recently IL-12, IFN-α, IFN-γ, and TLR antagonists have been shown to mediate the immune responses, specifically during chemotherapy against leishmaniasis [Citation158,Citation160]. The success of these adjuvants in traditional drug treatment could be translated into vaccine development to promote the induction of a strong Th1 cell-mediated response, crucial for protection and resistance to Leishmania infection [Citation161]. Another limitation to subunit vaccines is that isolation of enough purified protein is time consuming and difficult. Target molecules may also become misfolded, or errors may occur in the post-translational processing, transport, and localization process of the protein. However, subunit vaccines can also be advantageous since they do not include live component. Therefore, they pose no risk of infection and are suitable for immunocompromised individuals.

Interestingly, investigations involving Leishmania sand fly vector saliva and promastigote secretory gel (PSG) have revealed its impact on modulating the bite site immune environment and development of disease [Citation162–164]. Leishmania parasites have shown increased virulence when transmitted by sand fly vector, this virulence being associated with proteins in the saliva of the New World vector species Lutzomyia longipalpis [Citation165,Citation166]. Vector-derived factors of transmission are rarely considered in vaccine development, however, due to these observations, sand fly salivary proteins have been investigated as potential vaccine candidates either alone or combined with Leishmania proteins. It has been reported that individual saliva antigens are able to elicit paradoxical effects on disease outcome than the whole saliva. The three most promising candidates to date are LJM19 and LJL143 from the L. infantum vector Lutzomyia longipalpis, and PdSP15 from the L. major vector P. duboscqui. It is hypothesized that these proteins normally function to recruit phagocytic macrophages and neutrophils to the bite site by increasing vasodilation, in turn promoting the infection. However, studies of these candidates on mice, hamsters, dogs, and non-human primates revealed their immunogenicity and effectiveness in protecting against infection [Citation167–172]. Furthermore, salivary proteins have been shown to induce the cell-mediated immune response without the addition of an adjuvant, unlike recombinant protein candidates [Citation170]. Thus, investigation of sand fly salivary proteins in Leishmania vaccine development continues to be a growing area of the field.

5.3. DNA vaccines

More recently, genetic immunization strategies have arisen as third generation vaccines that utilize direct injection of nucleic acids, either as mRNA, naked plasmid DNA, or encapsulated in a viral vector. An RNA vaccine expressing LEISH-F2 followed by a subunit vaccine was shown to protect against L. donovani in murine models [Citation173]. Furthermore, interest in DNA vaccines began after observation that intramuscular injection of plasmid DNA stimulated the production of its encoded protein in vivo [Citation174]. In the case of Leishmania, a large repertoire of antigens have been investigated, with the most prevalent DNA vaccines encoding proteins previously tested as recombinant vaccine candidates [Citation148]. Such candidates include single vaccines consisting of one or more protein genes fused on a single plasmid [Citation175–181], or multigenic vaccines possessing multiple protein genes in a cocktail of separate plasmids [Citation178,Citation182–185]. These vaccines remain in pre-clinical trials and have been tested against the CL strains of L. major, L. mexicana, and L. amazonensis in mice, as well as the VL strains of L. donovani, L. infantum, and L. chagasi in mice, hamster, and dog models [Citation148].

The first DNA vaccine developed for leishmaniasis was composed of the gene encoding gp63, which has been shown to induce a Th1 response and significantly protect against L. major infection [Citation177,Citation186,Citation187] and partially protect against L. mexicana infection in BALB/c mice [Citation188,Citation189]. Another DNA vaccine known as LACK has been the most extensively studied Leishmania DNA vaccine, delivering various degrees of protection in cutaneous (L. major) and visceral (L. donovani and L. infantum) infections in mouse and dog models [Citation175,Citation190–193]. Additional DNA vaccine candidates that have induced protective immunity in animal models include genes encoding for proteins such TSA, KMP11, PSA-2, A-2, NH36, LmSTI1, cysteine proteases, and histones and have been previously reviewed [Citation148,Citation149,Citation194]. Heterologous prime boosting has also been utilized to increase the immunogenicity of these vaccines [Citation195–197]. In this method, the DNA vaccine is first used to prime, followed by a booster of the corresponding protein expressed on a recombinant viral vector such as vaccinia virus (VV) or modified vaccinia virus Ankara (MVA). Vaccine efficacy can thus be improved by enhancing the humoral and cell-mediated responses, as well as increasing the production of protective cytokines [Citation198].

DNA vaccines are appealing due to their ease of production, stability, and safety. They are also known to elicit a stronger Th1 immune responses than their protein vaccine counterparts [Citation199]. Expression of the encoded antigen occurs over the course of several days and allows for induction of a humoral and cell-mediated response leading to long-term immunity. The fusion of multiple antigen genes onto a single plasmid is an additional advantage of DNA vaccines. Furthermore, naked DNA vaccines lacking a viral coat are resistant against neutralizing antibody reactions toward viral coat proteins, a problem observed with recombinant vaccines. On the other hand, the lack of this protein coat remains as a disadvantage to naked DNA vaccines since it usually serves to protect against DNA degradation and aiding delivery to host cells [Citation200]. Overall, DNA vaccines have been successful in animal models, although, translation to human has been more difficult owing to their poor immunogenicity and inability to generate significant clinical benefits. Thus, there is currently no DNA vaccine approved for human use not only for leishmaniasis but also against other infections [Citation201]. However, optimization strategies continue to be investigated, such as optimization of transfection and the use of a prime-boost strategy that has improved the efficacy in larger animals and humans [Citation194,Citation201].

5.4. Live-attenuated vaccines

Live-attenuated vaccines involve the genetic deletion of genes responsible for the virulence and/or survival of an organism. They are able to induce an immune response like that of a natural infection without the risk of disease. The live parasite is properly delivered and processed through its natural infection pathway, and all parasite antigens remain expressed for presentation to the host immune system. This allows for the development of specific effector cells and long-term protective immunity against reinfection [Citation202]. Attenuation can be accomplished by two different methods: undefined attenuation from long-term in vitro culture by using irradiation, chemical mutagenesis, or selective pressure, or alternatively, defined attenuation by specifically removing a target virulence gene [Citation203]. One of the historical ways of disrupting Leishmania virulence genes is by homologous recombination of the two alleles with selectable marker genes [Citation204]. Suicide cassettes have also been utilized to introduce drug sensitive genes into the Leishmania genome to control infection and induce immunity [Citation205–207].

In the case of Leishmania, the availability of the complete Leishmania genome sequence has facilitated the effort to identify and analyze genes involved in virulence of the parasite to produce defined vaccine candidates [Citation150]. Multiple defined live-attenuated vaccines generated from L. major, L. mexicana, L. amazonensis, and L. donovani have been developed that display significant protection against CL and VL in susceptible mice [Citation208–212]. In particular, Biopterin transporter 1 (BT1)-deleted L. donovani parasites have been shown to be less infective than their WT counterpart, and to promote protective immunity characterized by IFN-γ. However, BT1 null parasites were still detected in the spleen and liver after 4 weeks of injection, and could pose safety risk [Citation213]. Another L. donovani genetically modified strain was generated by deleting the gene coding for Ldp27, a protein of the cytochrome c complex abundantly expressed in amastigotes. Immunization with Ldp27-deficient parasites resulted in reduced parasitic loads and enhanced Th1 long term protection in CL and VL challenge models [Citation214,Citation215]. Another group created a heat shock protein (HSP)-70-II null L. infantum vaccine candidate. This strain also showed lower parasitic burdens and enhanced protective immunity against a model of CL, but not VL [Citation216–219]. Furthermore, Santi, et al. showed that L. infantum KHARON1 (KH1) null mutants present growth defects and are unable to infect macrophages in vitro, however long-lasting protection has still not been demonstrated [Citation220]. In general, many gene targets have been the focus of defined attenuated Leishmania vaccines, such as dhfr, lpg2, cpa, cpb, Ufm1, p27, SIR2, BT1, HSP70, centrin, and the paraflagellar rod-2 locus which are reviewed in these references [Citation148,Citation149,Citation202,Citation203]. However, evaluation of these genetically modified candidates in animal models has exhibited mixed results of protection.

Safety is of paramount importance to vaccine development. Although live-attenuated vaccines best mimic natural infection and induce protective immunity, safety constraints limit their advantage. Potential reversion to a wildtype virulent form, presence of the antibiotic resistant genes or the risk associated with vaccinating immunosuppressed individuals are relevant concerns [Citation149]. However, advances in genetic manipulation such as the CRISPR/Cas9 system can overcome some of these barriers. Utilizing CRISPR/Cas9 technology to generate attenuated Leishmania vaccine candidates is advantageous since it does not require the use of antibiotic-resistance selection genes, a requirement for advancement to human trials. Vaccines generated in this manner, namely the centrin-deleted (L. donovani Centrin-/- was not generated with the CRISPR/Cas9 technique) L. major vaccine candidate, has proven to be safe, immunogenic, and protective in mice and hamsters [Citation118,Citation222]. Furthermore, it has also been shown that centrin-deleted L. major parasites do not survive inside the sand fly gut. This removes the risk of attenuated parasites being picked up by the sand fly and regaining virulence through recombination with wild-type parasites in the sand fly gut [Citation222]. Thus, live-attenuated Leishmania vaccines have a promising future in the field.

A subset of the above-mentioned vaccines is shown in ; exemplifying the advantages and disadvantages of different vaccination methods.

Table 1. Diverse vaccination approaches tested against Leishmania

6. Common challenges in the development of a Leishmania vaccine

In the absence of a vaccine, natural Leishmania infection is currently the main immunization process, as cohort population studies show that recovered patients are usually protected for life [Citation223,Citation224]. These patients also display a positive response to a leishmanin skin test (LST). LST can be used as a diagnostic method to identify previous Leishmania infections, as it consists of the development of a delayed-type hypersensitivity (DTH) skin reaction in response to intradermally injected Leishmania antigen. LST elicits swelling, inflammation and induration, accompanied by a strong Th1 and macrophage response in recovered patients of CL and VL, even after many years after the infection, which exemplifies how natural infections act as a method of efficient immunizations [Citation225–228]. A more in-depth discussion about the relevance of LST can be found at [Citation229].

These data have encouraged some countries, such as Israel, Iran, and Uzbekistan to use the inoculation of live L. major, a skin restricted strain, as a vaccination method until recent years. The intentional intradermal immunization in covered parts of the body with live parasites is called Leishmanization, and it has been successful in reducing the incidence of CL in target countries [Citation230]. Nevertheless, as we learnt from the experience of Iran, besides its effectiveness, leishmanization might result in the development of chronic or temporary non-healing lesions, and there is also the risk of culture contamination with fungi or bacteria [Citation231,Citation232]. Even when there is a reduction of CL incidence, leishmanization does not eliminate the problem because infected individuals are also reservoirs for the parasite and contribute to L. major transmission [Citation233]. So far, there is not an equivalent immunization process against VL or MCL, since both manifestations of leishmaniasis lead to aggressive symptoms, and in the case of VL, even death. For this reason, it is not feasible to use leishmanization for MCL or VL.

The success of Leishmanization and natural Leishmania infection in the development of protection against CL proves that vaccination is possible. As shown in the previous section, many vaccine candidates have been proposed so far, nevertheless, there are still some challenges related to the biology of the infection and the manufacture which need to be overcome before achieving a safe and efficacious vaccine.

First, there is a concern about the balance between safety and efficacy. Live attenuated vaccines can achieve attenuation by either physicochemical methods (i.e. long-term invitro culture, mutagenesis, or irradiation) or by genetic modifications. In the first case, there is the concern that the parasites could regain virulence; this is also possible for parasites with mutations in just one allele of the genes [Citation203]. Laboratory-generated Leishmania mutants are also antibiotic resistant, which is a useful and practical method for parasite selection during scientific research. Some parasites may retain those genes, which can also lead to antibiotic resistance within Leishmania populations, making this type of vaccine not feasible for clinical use [Citation234]. Thus, homozygous mutants without antibiotic resistance genes would be ideal candidates for safe use in humans. Additionally, live attenuated vaccines should be designed to be safe for immunocompromised patients, since vaccination is also necessary in regions with high rates of HIV-positive individuals [Citation203]. On that regard, killed parasites may appear as a safer alternative, providing a broad range of antigens without the risk of virulence. However, they fail to provide significant protection, suggesting that a carefully designed live attenuated vaccine could be a better option [Citation131].

Another factor to consider when testing the efficacy of a novel vaccine is the difference between needle and sand fly challenge [Citation235]. The sand fly saliva contains molecules which facilitate the bloodmeal by dampening coagulation or modulate the host responses, either favoring immune evasion or inducing an immune response [Citation42,Citation236]. Differences in the protection, immune profile and time of response have been observed when immunized mice were either challenged by sand fly bite or needle injection; and the immune response also changed depending on the initial immunization (live versus killed L. major) [Citation237,Citation238]. Even in experimental models of sand fly infection, the dose of parasites delivered and the frequency varies within populations, making the vaccine testing process more complex [Citation239]. Therefore, there is a critical need for testing candidate vaccines in animal models of sand fly challenge, as it more closely resembles clinical infection.

Finally, the fact that there are many different clinical manifestations and immune responses against the variety of Leishmania species, makes the development of a single vaccine protective against all the different species even more complicated [Citation129,Citation203].

It is noteworthy to mention that there is also an historical delay in the development of vaccines against NTD, including leishmaniasis. This could be due to the fact that vaccines are less profitable than drugs, hence, they are an unattractive market for industry [Citation129,Citation240].

Once the complications of developing a safe and efficacious vaccine can be overcome, the clinical trials need to be properly designed. This might not be a straightforward decision as identifying the right region and population has its own issues. For example, even in Bihar, the Indian state with the highest incidence VL rate, there are areas in the state which reported less than 1 case per 10,000. This incidence is too low to perform a clinical phase 3 including a group receiving the in-test vaccine and another receiving an alternative immunization [Citation229]. Performing clinical trials in endemic regions is necessary to test the protection against Leishmania after challenge, nevertheless, this means that participants will be exposed to the sand flies for uncontrolled times, resulting in differences in the parasite inoculation [Citation230].

7. Concluding remarks

Leishmaniasis affects more than 88 countries around the globe, many of which are low to middle income countries (LMIC). Despite the high morbidity and mortality of this disease there are no vaccines for human use currently available. So far, many candidate vaccines, including killed or live-attenuated parasites, recombinant proteins, and DNA vaccines have been developed [Citation129]. However, challenges such as safety, efficacy, the selection of adjuvants, and the need for sand fly challenge testing remain. Leishmanization has been shown to be the most effective vaccination against CL. CRISPR-based methods enabled the development of Leishmania genetic mutants with precise deletions that embody the immunogenicity and protection effects observed in virulent L. major parasites, but without causing pathology [Citation118,Citation221]. Preclinical studies have shown that genetically modified live attenuated Leishmania parasites can also confer protection against VL, raising the prospect of a safe and efficacious pan-Leishmania vaccine for the first time [Citation222,Citation241].

Toward realizing this goal, several considerations must be satisfied. First, we need to collaborate and incentivize industry partners to manufacture vaccines under Good Manufacturing Practices (cGMP) conditions since such vaccines are not profitable. Secondly, a typical Phase III trial is likely to be prohibitively expensive due to the necessity to include large number of subjects, only a small fraction of which are likely to develop the disease following natural exposure. To mitigate this problem, alternative vaccine trial models, such as a Challenge Human Infection Model (CHIM), can be employed as previously shown in other disease models [Citation242]. However careful and stringent incorporation of exploratory biomarkers, as well as the reintroduction of a standardized LST as a surrogate biomarker of immunogenicity, in Phase I studies should also be investigated to demonstrate vaccine safety and immunogenicity both in endemic and non-endemic countries. This will help to proceed to Phase III efficacy trials. Lastly, we need a vaccine which is affordable for LMICs. Despite its significant impact on public health and the economy of endemic countries, the global impact of leishmaniasis is often ignored by the international community because it is the disease of the poor [Citation80].

8. Expert opinion

Leishmaniasis is one of the top ten Neglected Tropical Diseases and impacts large populations in LMICs. With the increased migrations from and tourism to LMICs, this disease has the potential to spread in non-endemic countries. Little is known about the global impact of this disease, pathogenesis, and treatment options. In addition, there is a lack of effective strategies to control this disease, a need for new diagnostic assay development approaches such as involving omic approaches, and a need for discovery of new drugs that would not result in parasite resistance. On the other hand, vaccination can be one of the effective measures to control and/or eliminate leishmaniasis. Previously, several efforts have been made to develop vaccines against various species of Leishmania. However, very few of the candidate vaccines have been rigorously evaluated beyond pre-clinical studies to assess safety, efficacy, select dose and adjuvants, and to satisfy the need for natural sand fly challenge essential for progress toward clinical studies and regulatory approval. It has been demonstrated that people who have recovered from leishmaniasis are protected for lifetime from future infections, a practice come to be known as Leishmanization. Leishmanization has been widely considered be the most effective vaccination against CL. However, because of the uncertain safety of this practice, it is no longer practiced widely. Recently, genetically modified Leishmania parasites as prophylactic vaccines have been tested in the pre-clinical models that showed protection similar to leishmanization. Therefore, to fully realize the potential of Leishmania parasites as safe and efficacious vaccines, several considerations must be satisfied. First and foremost, a full characterization of the vaccine strain is necessary to meet the expected standards of safety by the regulatory authorities. This includes manufacture of the vaccine strain under cGMP conditions with complete genome sequences, lot to lot consistency, well-defined growth conditions, and absence of adventitious agents toward meeting the safety, identity, purity, and potency standards. For example, some species of Leishmania parasites (both new and old world) contain endogenous Leishmania RNA viruses, which are known to promote disease exacerbation [Citation243]. In addition, care must be taken to develop a media formulation that is free from serum containing Bovine Spongiform Encephalopathy agent. In some situations, it may be necessary to avoid use of serum altogether and use alternatives to serum because of the religious beliefs and cultural norms prevalent in some of the endemic countries. Secondly, there needs to be a well-designed plan to assess efficacy of such vaccines in endemic countries. For example, in many endemic countries there are other co-infections along with Leishmania, therefore the vaccine of choice should be efficacious under these conditions. The choice of clinical trial sites must be such that there is significant incidence and prevalence of the disease since there are variations in the incidence in the subregions of the endemic areas. This may require a large number of subjects, which may not be possible in some situations and may take years to complete the efficacy studies due to the considerable lag between exposure and development of acute disease. Recently, an alternative model of vaccine efficacy known as Challenge Human Infection Model (CHIM) is being tested for various pathogens. Vaccine developers need to explore CHIM to test Leishmania vaccine efficacy. In addition, there is a need for well defined correlates of protection. Toward that goal there is a need to explore new biomarkers of immunogenicity in addition to the markers widely used in preclinical studies. For example, one should explore the well studied Leishmania Skin Test (LST) as a surrogate biomarker of immunogenicity. However, LST is not currently being produced under cGMP conditions that would be permissible in clinical use. Similarly, use of new upcoming technologies, such as single cell RNA seq and/or NanoString RNA and DNA analysis, will help identify new biomarkers of immunogenicity in the exploratory investigations early in the clinical studies, which could be correlated with potential protection. Since Leishmania parasites cause a wide spectrum of diseases, and manufacturers generally lack market incentives to make vaccines against leishmaniasis, it may be desirable to develop a pan-Leishmania vaccine that may be efficacious against multiple species of the parasite. Live attenuated L. major parasites (LmCen−/-) have shown early promise in affording protection against heterologous challenge with L. donovani indicating the feasibility of a pan-Leishmania vaccine [Citation222]. Finally, we need to develop partnerships with the vaccine manufacturers and create an ongoing dialogue with the authorities in the LMICs to highlight the importance of a Leishmania vaccine and its potential in control and elimination of leishmaniasis and to achieve global public health goals.

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. The statements made in this review represent the FDA authors' own best judgments. These comments do not bind or obligate the Food and Drug Administration.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was funded by National Institutes of Health, Global Health Innovation Technology Fund.

References

  • Organization WH. Investing to overcome the global impact of neglected tropical diseases: third WHO report on neglected tropical diseases (World Health Organization, Geneva, 2015).
  • Maroli M, Feliciangeli Md, Bichaud L, et al. Phlebotomine sandflies and the spreading of leishmaniases and other diseases of public health concern. Med Vet Entomol. 2013;27(2):123–147.
  • Kima PE. The amastigote forms of Leishmania are experts at exploiting host cell processes to establish infection and persist. Int J Parasitol. 2007;37(10):1087–1096.
  • Desjeux P. Leishmaniasis: current situation and new perspectives. Comp Immunol Microbiol Infect Dis. 2004;27(5):305–318.
  • Wilhelm TJ,Visceral leishmaniasis. Der Chirurg. 2019;90(10):833–837.
  • Pennisi MG, Persichetti MF. Feline leishmaniosis: is the cat a small dog? Vet Parasitol. 2018;251:131–137.
  • Mhadhbi M, Sassi A. Infection of the equine population by Leishmania parasites. Equine Vet J. 2020;52(1):28–33.
  • Han S, Wu W-P, Chen K, et al. Epidemiological survey of sheep as potential hosts for Leishmania in China. BMC Vet Res. 2018;14(1):378.
  • Pareyn M, Hendrickx R, Girma N, et al. Evaluation of a pan-Leishmania SL RNA qPCR assay for parasite detection in laboratory-reared and field-collected sand flies and reservoir hosts. Parasit Vectors. 2020;13(1). DOI:https://doi.org/10.1186/s13071-020-04141-y.
  • Chen H, Li J, Zhang J, et al. Multi-locus characterization and phylogenetic inference of Leishmania spp. in snakes from Northwest China. PLoS ONE. 2019;14(4).
  • Zhang J-R, Guo X-G, Liu J-L, et al. Molecular detection, identification and phylogenetic inference of Leishmania spp. in some desert lizards from Northwest China by using internal transcribed spacer 1 (ITS1) sequences. Acta Trop. 2016;162:83–94.
  • Ashford RW. Leishmaniasis reservoirs and their significance in control. Clin Dermatol. 1996;14(5):523–532.
  • Tomassone L, Berriatua E, De Sousa R, et al. Neglected vector-borne zoonoses in Europe: into the wild. Vet Parasitol. 2018;251:17–26.
  • Tabbabi A. Review of Leishmaniasis in the Middle East and North Africa. Afr Health Sci. 2019;19(1):1329–1337.
  • Ghawar W, Bettaieb J, Salem S, et al. Natural infection of Ctenodactylus gundi by Leishmania major in Tunisia. Acta Trop. 2018;177:89–93.
  • Franco AO, Davies CR, Mylne A, et al. Predicting the distribution of canine leishmaniasis in western Europe based on environmental variables. Parasitology. 2011;138(14):1878–1891.
  • Miguel DC, Gaurnier DC. Canine and Human Leishmaniasis: disease progression to Brazilian Urbanized areas. 2019 2(3); International Journal of Tropical Diseases.
  • Dantas-Torres F, Brandão-Filho SP. Visceral leishmaniasis in Brazil: revisiting paradigms of epidemiology and control. Rev Inst Med Trop Sao Paulo. 2006;48(3):151–156.
  • Gurel MS, Tekin B, Uzun S. Cutaneous leishmaniasis: a great imitator. Clin Dermatol. 2020;38(2):140–151.
  • Akilov O, Khachemoune A, Hasan T. Clinical manifestations and classification of old World cutaneous leishmaniasis. Int J Dermatol. 2007;46(2):132–142.
  • Volpedo G, Pacheco-Fernandez T, Holcomb EA, et al. Mechanisms of immunopathogenesis in Cutaneous Leishmaniasis and Post Kala-azar Dermal Leishmaniasis (PKDL). Front Cell Infect Microbiol. 2021;11. DOI:https://doi.org/10.3389/fcimb.2021.685296
  • Handler MZ, Patel PA, Kapila R, et al. Cutaneous and mucocutaneous leishmaniasis: differential diagnosis, diagnosis, histopathology, and management. J Am Acad Dermatol. 2015;73(6):911–926.
  • Srivastava P, Dayama A, Mehrotra S, et al. Diagnosis of visceral leishmaniasis. Trans R Soc Trop Med Hyg. 2011;105(1):1–6.
  • Vega-López F. Diagnosis of cutaneous leishmaniasis. Curr Opin Infect Dis. 2003;16(2):97–101.
  • Sundar S, Singh OP. Molecular diagnosis of Visceral Leishmaniasis. Mol Diagn Ther. 2018;22(4):443–457.
  • Thakur S, Joshi J, Kaur S. Leishmaniasis diagnosis: an update on the use of parasitological, immunological and molecular methods. J Parasitic Dis. 2020;44(2):253–272.
  • Van Griensven J, Visceral Leishmaniasis: DE. Recent advances in diagnostics and treatment regimens. Infect Dis Clin North Am. 2019;33(1):79–99.
  • Moreira OC, Yadon ZE, Cupolillo E. The applicability of real-time PCR in the diagnostic of cutaneous leishmaniasis and parasite quantification for clinical management: current status and perspectives. Acta Trop. 2018;184:29–37.
  • Aronson NE, Joya CA, LeishmaniasisCutaneous Leishmaniasis: Updates in Diagnosis and Management. Infect Dis Clin North Am. 2019;33(1):101–117.
  • Rosenthal E, Marty P. Recent understanding in the treatment of visceral leishmaniasis. J Postgrad Med. 2003;49(1):61.
  • Sundar S, More DK, Singh MK, et al. Failure of Pentavalent Antimony in Visceral Leishmaniasis in India: report from the center of the Indian Epidemic. Clinl Infect Dis. 2000;31(4):1104–1107.
  • World Health Organization. Cutaneous Leishmaniasis control in selected countries of the two eastern Mediterranean and African regions. 2015. Geneva, Switzerland: World Health Organization
  • Shirzadi MR. Lipsosomal amphotericin B: a review of its properties, function, and use for treatment of cutaneous leishmaniasis. Res Rep Trop Med. 2019;10:11–18.
  • Azab AS, Kamal MS, Haggar MSE, et al. Early surgical treatment of Cutaneous Leishmaniasis. The Journal of Dermatologic Surgery and Oncology. 1983;9(12):1007–1012.
  • Bassiouny A, Meshad ME, Talaat M, et al. Cryosurgery in cutaneous leishmaniasis. Br J Dermatol. 1982;107(4):467–474.
  • Panagiotopoulos A, Stavropoulos PG, Hasapi V, et al. Treatment of cutaneous leishmaniasis with cryosurgery. Int J Dermatol. 2005;44(9):749–752.
  • Aronson NE, Wortmann GW, Byrne WR, et al. A randomized controlled trial of local heat therapy versus intravenous sodium stibogluconate for the treatment of cutaneous Leishmania major Infection. PLoS Negl Trop Dis. 2010;4(3):e628.
  • Bumb RA, Satoskar AR. Radiofrequency-induced heat therapy as first-line treatment for cutaneous leishmaniasis. Expert Rev Anti Infect Ther. 2011;9(6):623–625.
  • David JR. The successful use of radiofrequency-induced heat therapy for cutaneous leishmaniasis: a review. Parasitology. 2018;145(4):527–536.
  • Frolich S, Kaplan I. Plastic surgery in the treatment of Cutaneous Leishmaniasis of the face. Plast Reconstr Surg. 1967;40(2):157–160.
  • Rodrigues V, Cordeiro-da-Silva A, Laforge M, et al. Regulation of immunity during visceral Leishmania infection. Parasit Vectors. 2016;9:118. DOI:https://doi.org/10.1186/s13071-016-1412-x
  • Pacheco-Fernandez T, Volpedo G, Verma C, et al. Understanding the immune responses involved in mediating protection or immunopathology during leishmaniasis. Biochem Soc Trans. 2021;49(1):297–311.
  • Nylén S, Gautam S. Immunological perspectives of leishmaniasis. J Glob Infect Dis. 2010;2(2):135–146.
  • Sharma U, Singh S. Immunobiology of leishmaniasis. In: IJEB Vol.47(06) [June 2009]. Indian Journal of Experimental Biology. 2009.
  • Kaye PM, Aebischer T. Visceral leishmaniasis: immunology and prospects for a vaccine. Clin Microbiol Infect. 2011;17(10):1462–1470.
  • Kaye PM, Svensson M, Ato M, et al. The immunopathology of experimental visceral leishmaniasis. Immunol Rev. 2004;201(1):239–253.
  • Scott P, Novais FO. Cutaneous leishmaniasis: immune responses in protection and pathogenesis. Nat Rev Immunol. 2016;16(9):581–592.
  • Real F, Florentino PTV, Reis LC, et al. Cell-to-cell transfer of Leishmania amazonensis amastigotes is mediated by immunomodulatory LAMP-rich parasitophorous extrusions. Cell Microbiol. 2014;16(10):1549–1564.
  • Hohman LS, Peters NC. CD4+ T Cell-Mediated Immunity against the Phagosomal Pathogen Leishmania: implications for Vaccination. Trends Parasitol. 2019;35(6):423–435.
  • Glennie ND, Volk SW, Skin-resident SP. CD4+ T cells protect against Leishmania major by recruiting and activating inflammatory monocytes. PLoS Pathog. 2017;13(4):e1006349.
  • Qadoumi M, Becker I, Donhauser N, et al. Expression of inducible nitric oxide synthase in skin lesions of patients with American Cutaneous Leishmaniasis | infection and immunity. ASM Journals Infection and Immunity. 2020 70(8).
  • Á G, Valério-Bolas A, Palma-Marques J, et al. Cutaneous Leishmaniasis: the complexity of host’s effective immune response against a polymorphic parasitic disease. J Immunol Res. 2019;2019.
  • Bhor R, Rafati S, Pai K. Cytokine saga in visceral leishmaniasis. Cytokine. 2020;155322. DOI:https://doi.org/10.1016/j.cyto.2020.155322
  • Alexander J, Carter KC, Al-Fasi N, et al. IL-4 is necessary for effective drug therapy against visceral leishmaniasis. Eur J Immunol. 2000;30(10):2935–2943.
  • Goto H, Lindoso J. Immunity and immunosuppression in experimental visceral leishmaniasis. Braz J Med Biol Res. 2004;37(4):615–623.
  • Rossi M, Fasel N. How to master the host immune system? Leishmania parasites have the solutions! Int Immunol. 2017;30(3):103–111.
  • Silveira FT, Lainson R, De Castro Gomes CM, et al. Immunopathogenic competences of Leishmania (V.) braziliensis and L. (L.) amazonensis in American cutaneous leishmaniasis. Parasite Immunol. 2009;31(8):423–431.
  • Srivastava P, Gidwani K, Picado A, et al. Molecular and serological markers of Leishmania donovani infection in healthy individuals from endemic areas of Bihar, India. Trop Med Int Health. 2013;18(5):548–554.
  • Nweze JA, Nweze EI, Onoja US. Nutrition, malnutrition, and leishmaniasis. Nutrition. 2020;73:110712.
  • Akuffo H, Costa C, Van Griensven J, et al. New insights into leishmaniasis in the immunosuppressed. PLoS Negl Trop Dis. 2018;12(5):e0006375.
  • Sakthianandeswaren A, Foote SJ, Handman E. The role of host genetics in leishmaniasis. Trends Parasitol. 2009;25(8):383–391.
  • Van Griensven J, Carrillo E, López-Vélez R, et al. Leishmaniasis in immunosuppressed individuals. Clin Microbiol Infect. 2014;20(4):286–299.
  • Monge-Maillo B, Norman FF, Cruz I, et al. Visceral Leishmaniasis and HIV Coinfection in the mediterranean region. PLoS Negl Trop Dis. 2014;8(8):e3021. doi: https://doi.org/10.1371/journal.pntd.0003021
  • Malafaia G. Protein-energy malnutrition as a risk factor for visceral leishmaniasis: a review. Parasite Immunol. 2009;31(10):587–596.
  • Cuypers B, Berg M, Imamura H, et al. Integrated genomic and metabolomic profiling of ISC1, an emerging Leishmania donovani population in the Indian subcontinent. Infect Genet Evol. 2018;62:170–178.
  • Urrea DA, Duitama J, Imamura H, et al. Genomic analysis of Colombian Leishmania panamensis strains with different level of virulence. Sci Rep. 2018;8(1):17336.
  • Samarasinghe SR, Samaranayake N, Kariyawasam UL, et al. Genomic insights into virulence mechanisms of Leishmania donovani: evidence from an atypical strain. BMC Genomics. 2018;19(1):843.
  • Yasur-Landau D, Jaffe CL, David L, et al. Resistance of Leishmania infantum to allopurinol is associated with chromosome and gene copy number variations including decrease in the S-adenosylmethionine synthetase (METK) gene copy number. Int J Parasitol Drugs Drug Resist. 2018;8(3):403–410.
  • Sterkers Y, Crobu L, Lachaud L, et al. Parasexuality and mosaic aneuploidy in Leishmania: alternative genetics. Trends Parasitol. 2014;30(9):429–435.
  • Rougeron V, De Meeûs T, Bañuls AL. Reproduction in Leishmania: a focus on genetic exchange. Infect Genet Evol. 2017;50:128–132.
  • Telittchenko R, Descoteaux A. Study on the occurrence of genetic exchange among parasites of the Leishmania mexicana complex. Front Cell Infect Microbiol. 2020;10(607253). DOI:https://doi.org/10.3389/fcimb.2020.607253
  • Romano A, Inbar E, Debrabant A, et al. Cross-species genetic exchange between visceral and cutaneous strains of Leishmania in the sand fly vector. Proc Natl Acad Sci U S A. 2014;111(47):16808–16813.
  • World Health Organization. 2016. Weekly epidemiological record. (Ed.^(Eds). 421–432.
  • World Health Organization. Leishmaniasis, status of endemicity of cutaneous leishmaniasis: 2019. (Ed.^(Eds) (2019)
  • World Health Organization. Leishmaniasis. editor. Global health observatory data repository. 2016. Available from: https://apps.who.int/gho/data/node.main.NTDLEISH?lang=en
  • Instituto Ramon y Cajal de Investigacion Sanitaria, World Health Organization. 2019. WHO collaborating centre for clinical management of leishmaniasis (spa-55). (Ed.^(Eds).
  • World Health Organization W. 2015. Investing to overcome the global impact of neglected tropical diseases. (Ed.^(Eds).
  • Valero NNH, Uriarte M. Environmental and socioeconomic risk factors associated with visceral and cutaneous leishmaniasis: a systematic review. Parasitol Res. 2020;119(2):365–384.
  • Gadisa E, Tsegaw T, Abera A, et al. Eco-epidemiology of visceral leishmaniasis in Ethiopia. Parasit Vectors. 2015;8(1). DOI:https://doi.org/10.1186/s13071-015-0987-y.
  • Ngere I, Gufu Boru W, Isack A, et al. Burden and risk factors of cutaneous leishmaniasis in a peri-urban settlement in Kenya, 2016. PLoS ONE. 2020;15(1):e0227697. doi: https://doi.org/10.1371/journal.pone.0227697
  • Yadon ZE, Rodrigues LC, Davies CR, et al. Indoor and peridomestic transmission of American Cutaneous Leishmaniasis in Northwestern Argentina: a retrospective case-control study. Am J Trop Med Hyg. 2003;68(5):519–526.
  • Hotez PJ, Remme JHF, Buss P, et al. Combating tropical infectious diseases: report of the disease control priorities in developing countries project. Clinl Infect Dis. 2004;38(6):871–878.
  • McIlwee BE, Weis SE, Hosler GA. Incidence of endemic human cutaneous Leishmaniasis in the United States. JAMA Dermatol. 2018;154(9):1032.
  • Garrido-Jareño M, Sahuquillo-Torralba A, Chouman-Arcas R, et al. Cutaneous and mucocutaneous leishmaniasis: experience of a Mediterranean hospital. Parasit Vectors. 2020;13(1). DOI:https://doi.org/10.1186/s13071-020-3901-1.
  • Clarke CF, Bradley KK, Wright JH, et al. Emergence of Autochthonous Cutaneous Leishmaniasis in Northeastern texas and Southeastern Oklahoma. Am J Trop Med Hyg. 2013;88(1):157–161.
  • Curtin JM, Aronson NE. Leishmaniasis in the United States: emerging Issues in a Region of Low Endemicity. Microorganisms. 2021;9(3):578.
  • de Almeida M, Zheng Y, Nascimento FS, et al. Cutaneous Leishmaniasis caused by an unknown leishmania Strain, Arizona, USA. Emerg Infect Dis. 2021;27(6):1714–1717.
  • Bi K, Chen Y, Zhao S, et al. Current Visceral Leishmaniasis Research: a research review to inspire future study. Biomed Res Int. 2018;2018.
  • Hirve S, Kroeger A, Matlashewski G, et al. Towards elimination of visceral leishmaniasis in the Indian subcontinent—Translating research to practice to public health. PLoS Negl Trop Dis. 2017;11(10):e0005889.
  • World Health Organization. 2018. Leishmaniasis. (Ed.^(Eds)
  • Center for Disease Control and Prevention. 2013. Leishmaniasis. (Ed.^(Eds).
  • Okwor I, Social UJ. Economic burden of human Leishmaniasis. Am J Trop Med Hyg. 2016;94(3):489–493.
  • Kealey A, Smith R. Neglected tropical diseases: infection, modeling, and control. J Health Care Poor Underserved. 2010;21(1):53–69.
  • Hotez PJ,Ten failings in global neglected tropical diseases control. PLoS Negl Trop Dis. 2017;11(12):e0005896.
  • Brindha J,Balamurali M. M,and Kaushik Chanda. An overview on the therapeutics of neglected infectious diseases-Leishmaniasis and Chagas diseases. Front Chem. 2021;9:622286.
  • World Health Organization W. 2021. Neglected tropical diseases: treating more than one billion people for the fifth consecutive year. (Ed.^(Eds).
  • World Health Organization. 2020. Ending the neglect to attain the sustainable development goals. A road map for neglected tropical diseases 2021–2030. (Ed.^(Eds).
  • Ejov M, Dagne D. Strategic framework for leishmaniasis control in the WHO European Region 2014‒2020. (Ed.^(Eds) (2014)
  • Pan American Health Organization, World Health Organization. Plan of action to strengthen the surveillance and control of leishmaniasis in the Americas. (Ed.^(Eds) (2017)
  • Volpedo G, Costa L, Ryan N, et al. Nanoparticulate drug delivery systems for the treatment of neglected tropical protozoan diseases. J Venom Anim Toxins Incl Trop Dis. 2019;25:e144118.
  • Berry I, Berrang-Ford L. Leishmaniasis, conflict, and political terror: a spatio-temporal analysis. Soc Sci Med. 2016;167:140–149.
  • Pan American Health Organization Leishmaniasis P. (Ed.^(Eds)
  • Pavli A, Maltezou HC. Leishmaniasis, an emerging infection in travelers. Int J Infect Dis. 2010;14(12):e1032–1039.
  • Hamer DH, Rizwan A, Freedman DO, et al. GeoSentinel: past, present and future†. J Travel Med. 2020;27(8). DOI:https://doi.org/10.1093/jtm/taaa219
  • Koch LK, Kochmann J, Klimpel S, et al. Modeling the climatic suitability of leishmaniasis vector species in Europe. Sci Rep. 2017;7(1):13325.
  • Purse BV, Masante D, Golding N, et al. How will climate change pathways and mitigation options alter incidence of vector-borne diseases? A framework for leishmaniasis in South and Meso-America. PLoS One. 2017;12(10):e0183583.
  • Charrahy Z, Yaghoobi-Ershadi MR, Shirzadi MR, et al. Climate change and its effect on the vulnerability to zoonotic cutaneous leishmaniasis in Iran. Transbound Emerg Dis. 2021. DOI:https://doi.org/10.1111/tbed.14115.
  • Ready PD. Leishmaniasis emergence and climate change. Rev Sci Tech. 2008;27(2):399–412.
  • González C, Wang O, Strutz SE, et al. Climate change and risk of leishmaniasis in north america: predictions from ecological niche models of vector and reservoir species. PLoS Negl Trop Dis. 2010;4(1):e585.
  • Millán J, Ferroglio E, Solano-Gallego L. Role of wildlife in the epidemiology of Leishmania infantum infection in Europe. Parasitol Res. 2014;113(6):2005–2014.
  • Harhay MO, Olliaro PL, Costa DL, et al. Urban parasitology: visceral leishmaniasis in Brazil. Trends Parasitol. 2011;27(9):403–409.
  • Moreno J. Assessment of vaccine-induced immunity against Canine Visceral Leishmaniasis. Front Vet Sci. 2019;6:168.
  • Sousa-Paula LC, Silva LGD, Sales KGDS, et al. Failure of the dog culling strategy in controlling human visceral leishmaniasis in Brazil: a screening coverage issue? PLoS Negl Trop Dis. 2019;13(6):e0007553.
  • Dantas-Torres F, Miró G, Bowman DD, et al. Culling dogs for Zoonotic Visceral Leishmaniasis control: the wind of change. Trends Parasitol. 2019;35(2):97–101.
  • Ribeiro RR, Michalick MSM, Da Silva ME, et al. Canine Leishmaniasis: an overview of the current status and strategies for control. Biomed Res Int. 2018;2018:3296893.
  • Wagner V, Douanne N, Fernandez-Prada C. Leishmania infantum infection in a dog imported from Morocco. Can Vet J. 2020;61(9):963–965.
  • Velez R, Gállego M. Commercially approved vaccines for canine leishmaniosis: a review of available data on their safety and efficacy. Trop Med Int Health. 2020;25(5):540–557.
  • Zhang WW, Karmakar S, Gannavaram S, et al. A second generation leishmanization vaccine with a markerless attenuated Leishmania major strain using CRISPR gene editing. Nat Commun. 2020;11(1):3461.
  • Mendonça SC. Differences in immune responses against Leishmania induced by infection and by immunization with killed parasite antigen: implications for vaccine discovery. Parasit Vectors. 2016;9(1):492.
  • Saljoughian N, Taheri T, Rafati S. Live vaccination tactics: possible approaches for controlling visceral leishmaniasis. Front Immunol. 2014;5:134.
  • Le Rutte EA, Coffeng LE, Malvolti S, et al. The potential impact of human visceral leishmaniasis vaccines on population incidence. PLoS Negl Trop Dis. 2020;14(7):e0008468.
  • Michel G, Pomares C, Ferrua B, et al. Importance of worldwide asymptomatic carriers of Leishmania infantum (L. chagasi) in human. Acta Trop. 2011;119(2–3):69–75.
  • Molina R, Ghosh D, Carrillo E, et al. Infectivity of Post-Kala-azar Dermal Leishmaniasis patients to sand flies: revisiting a proof of concept in the context of the Kala-azar elimination program in the Indian subcontinent. Clin Infect Dis. 2017;65(1):150–153.
  • Asfaram S, Fakhar M, Mohebali M, et al. Asymptomatic human blood donors carriers of Leishmania infantum: potential reservoirs for visceral leishmaniasis in northwestern Iran. Transfus Apher Sci. 2017;56(3):474–479.
  • Singh OP, Tiwary P, Kushwaha AK, et al. Xenodiagnosis to evaluate the infectiousness of humans to sandflies in an area endemic for visceral leishmaniasis in Bihar, India: a transmission-dynamics study. Lancet Microbe. 2021;2(1):e23–e31.
  • Bacon KM, Hotez PJ, Kruchten SD, et al. The potential economic value of a cutaneous leishmaniasis vaccine in seven endemic countries in the Americas. Vaccine. 2013;31(3):480–486.
  • Lee BY, Bacon KM, Shah M, et al. The economic value of a visceral leishmaniasis vaccine in Bihar state, India. Am J Trop Med Hyg. 2012;86(3):417–425.
  • Cecílio P, Oristian J, Meneses C, et al. Engineering a vector-based pan-Leishmania vaccine for humans: proof of principle. Sci Rep. 2020;10(1):18653.
  • Srivastava S, Shankar P, Mishra J, et al. Possibilities and challenges for developing a successful vaccine for leishmaniasis. Parasit Vectors. 2016;9(1):277.
  • Modabber F. Vaccines against leishmaniasis. Ann Trop Med Parasitol. 1995;89(Suppl 1):83–88.
  • Noazin S, Modabber F, Khamesipour A, et al. First generation leishmaniasis vaccines: a review of field efficacy trials. Vaccine. 2008;26(52):6759–6767.
  • Noazin S, Khamesipour A, Moulton LH, et al. Efficacy of killed whole-parasite vaccines in the prevention of leishmaniasis: a meta-analysis. Vaccine. 2009;27(35):4747–4753.
  • Mayrink W, Williams P, Da Costa CA, et al. An experimental vaccine against American dermal leishmaniasis: experience in the State of Espírito Santo, Brazil. Ann Trop Med Parasitol. 1985;79(3):259–269.
  • Mayrink W, Da Costa CA, Magalhães PA, et al. A field trial of a vaccine against American dermal leishmaniasis. Trans R Soc Trop Med Hyg. 1979;73(4):385–387.
  • Vélez ID, del Pilar Agudelo S, Arbelaez MP, et al. Safety and immunogenicity of a killed Leishmania (L.) amazonensis vaccine against cutaneous leishmaniasis in Colombia: a randomized controlled trial. Trans R Soc Trop Med Hyg. 2000;94(6):698–703.
  • Sharples CE, Shaw MA, Castes M, et al. Immune response in healthy volunteers vaccinated with BCG plus killed leishmanial promastigotes: antibody responses to mycobacterial and leishmanial antigens. Vaccine. 1994;12(15):1402–1412.
  • Armijos RX, Weigel MM, Aviles H, et al. Field trial of a vaccine against New World cutaneous leishmaniasis in an at-risk child population: safety, immunogenicity, and efficacy during the first 12 months of follow-up. J Infect Dis. 1998;177(5):1352–1357.
  • Armijos RX, Weigel MM, Romero L, et al. Field trial of a vaccine against new world cutaneous leishmaniasis in an at-risk child population: how long does protection last? J Infect Dis. 2003;187(12):1959–1961.
  • Armijos RX, Weigel MM, Calvopina M, et al. Safety, immunogenecity, and efficacy of an autoclaved Leishmania amazonensis vaccine plus BCG adjuvant against New World cutaneous leishmaniasis. Vaccine. 2004;22(9–10):1320–1326.
  • Vélez ID, Gilchrist K, Arbelaez MP, et al. Failure of a killed Leishmania amazonensis vaccine against American cutaneous leishmaniasis in Colombia. Trans R Soc Trop Med Hyg. 2005;99(8):593–598.
  • Sharifi I, FeKri AR, Aflatonian MR, et al. Randomised vaccine trial of single dose of killed Leishmania major plus BCG against anthroponotic cutaneous leishmaniasis in Bam, Iran. Lancet. 1998;351(9115):1540–1543.
  • Alimohammadian MH, Khamesipour A, Darabi H, et al. The role of BCG in human immune responses induced by multiple injections of autoclaved Leishmania major as a candidate vaccine against leishmaniasis. Vaccine. 2002;21(3–4):174–180.
  • Bózner P, Gombosová A, Valent M, et al. Proteinases of Trichomonas vaginalis: antibody response in patients with urogenital trichomoniasis. Parasitology. 1992;105(Pt 3):387–391.
  • Khalil EA, El Hassan AM, Zijlstra EE, et al. Autoclaved Leishmania major vaccine for prevention of visceral leishmaniasis: a randomised, double-blind, BCG-controlled trial in Sudan. Lancet. 2000;356(9241):1565–1569.
  • Satti IN, Osman HY, Daifalla NS, et al. Immunogenicity and safety of autoclaved Leishmania major plus BCG vaccine in healthy Sudanese volunteers. Vaccine. 2001;19(15–16):2100–2106.
  • Momeni AZ, Jalayer T, Emamjomeh M, et al. A randomised, double-blind, controlled trial of a killed L. major vaccine plus BCG against zoonotic cutaneous leishmaniasis in Iran. Vaccine. 1999;17(5):466–472.
  • Mahmoodi M, Khamesipour A, Dowlati Y, et al. Immune response measured in human volunteers vaccinated with autoclaved Leishmania major vaccine mixed with low dose of BCG. Clin Exp Immunol. 2003;134(2):303–308.
  • Nagill R, Kaur S. Vaccine candidates for leishmaniasis: a review. Int Immunopharmacol. 2011;11(10):1464–1488.
  • Coutinho De Oliveira B, Duthie MS, Alves PVR. Vaccines for leishmaniasis and the implications of their development for American tegumentary leishmaniasis. Hum Vaccin Immunother. 2020;16(4):919–930.
  • Peacock CS, Seeger K, Harris D, et al. Comparative genomic analysis of three Leishmania species that cause diverse human disease. Nat Genet. 2007;39(7):839–847.
  • Kumari S, Kumar A, Samant M, et al. Discovery of novel vaccine candidates and drug targets against visceral leishmaniasis using proteomics and transcriptomics. Curr Drug Targets. 2008;9(11):938–947.
  • De Brito RCF, Cardoso JMO, Reis LES, et al. Peptide Vaccines for Leishmaniasis. Front Immunol. 2018;9:1043.
  • Alvar J, Croft SL, Kaye P, et al. Case study for a vaccine against leishmaniasis. Vaccine. 2013;31(Suppl 2):B244–249.
  • Singh B, Sundar S. Leishmaniasis: vaccine candidates and perspectives. Vaccine. 2012;30(26):3834–3842.
  • Gillespie PM, Beaumier CM, Strych U, et al. Status of vaccine research and development of vaccines for leishmaniasis. Vaccine. 2016;34(26):2992–2995.
  • Bertholet S, Goto Y, Carter L, et al. Optimized subunit vaccine protects against experimental leishmaniasis. Vaccine. 2009;27(50):7036–7045.
  • Coler RN, Duthie MS, Hofmeyer KA, et al. From mouse to man: safety, immunogenicity and efficacy of a candidate leishmaniasis vaccine LEISH-F3+GLA-SE. Clin Transl Immunology. 2015;4(4):e35.
  • Raman VS, Duthie MS, Fox CB, et al. Adjuvants for Leishmania vaccines: from models to clinical application. Front Immunol. 2012;3:144.
  • Askarizadeh A, Jaafari MR, Khamesipour A, et al. Liposomal adjuvant development for leishmaniasis vaccines. Ther Adv Vaccines. 2017;5(4–5):85–101.
  • Vogel FR, Hem SL. Chapter 5 - Immunologic adjuvants. In: Vaccines. (editor) Elsevier;2008. p. 59–71.
  • Carvalho EM, Johnson WD, Barreto E, et al. Cell mediated immunity in American cutaneous and mucosal leishmaniasis. J Immunol. 1985;135(6):4144–4148.
  • Rogers ME. The role of leishmania proteophosphoglycans in sand fly transmission and infection of the Mammalian host. Front Microbiol. 2012;3:223.
  • Abdeladhim M, Kamhawi S, Valenzuela JG. What’s behind a sand fly bite? The profound effect of sand fly saliva on host hemostasis, inflammation and immunity. Infect Genet Evol. 2014;28:691–703.
  • Gomes R, Oliveira F. The immune response to sand fly salivary proteins and its influence on leishmania immunity. Front Immunol. 2012;3:110.
  • Giraud E, Svobodová M, Müller I, et al. Promastigote secretory gel from natural and unnatural sand fly vectors exacerbate. Parasitology. 2019;146(14):1796–1802.
  • Martin-Martin I, Chagas AC, Guimaraes-Costa AB, et al. Immunity to LuloHya and Lundep, the salivary spreading factors from Lutzomyia longipalpis, protects against Leishmania major infection. PLoS Pathog. 2018;14(5):e1007006.
  • Gomes R, Teixeira C, Teixeira MJ, et al. Immunity to a salivary protein of a sand fly vector protects against the fatal outcome of visceral leishmaniasis in a hamster model. Proc Natl Acad Sci U S A. 2008;105(22):7845–7850.
  • Oliveira F, Rowton E, Aslan H, et al. A sand fly salivary protein vaccine shows efficacy against vector-transmitted cutaneous leishmaniasis in nonhuman primates. Sci Transl Med. 2015;7(290):290ra290.
  • Tavares NM, Silva RA, Costa DJ, et al. Lutzomyia longipalpis saliva or salivary protein LJM19 protects against Leishmania braziliensis and the saliva of its vector, Lutzomyia intermedia. PLoS Negl Trop Dis. 2011;5(5):e1169.
  • Gomes R, Oliveira F, Teixeira C, et al. Immunity to sand fly salivary protein LJM11 modulates host response to vector-transmitted leishmania conferring ulcer-free protection. J Invest Dermatol. 2012;132(12):2735–2743.
  • Collin N, Gomes R, Teixeira C, et al. Sand fly salivary proteins induce strong cellular immunity in a natural reservoir of visceral leishmaniasis with adverse consequences for Leishmania. PLoS Pathog. 2009;5(5):e1000441.
  • Reed SG, Coler RN, Mondal D, et al. Leishmania vaccine development: exploiting the host-vector-parasite interface. Expert Rev Vaccines. 2016;15(1):81–90.
  • Duthie MS, Van Hoeven N, MacMillen Z, et al. Heterologous immunization with defined RNA and subunit vaccines enhances T cell responses that protect against. Front Immunol. 2018;9(2420). DOI:https://doi.org/10.3389/fimmu.2018.02420.
  • Liu MA, Wahren B, Karlsson Hedestam GB. DNA vaccines: recent developments and future possibilities. Hum Gene Ther. 2006;17(11):1051–1061.
  • Gurunathan S, Sacks DL, Brown DR, et al. Vaccination with DNA encoding the immunodominant LACK parasite antigen confers protective immunity to mice infected with Leishmania major. J Exp Med. 1997;186(7):1137–1147.
  • Solioz N, Blum-Tirouvanziam U, Jacquet R, et al. The protective capacities of histone H1 against experimental murine cutaneous leishmaniasis. Vaccine. 1999;18(9–10):850–859.
  • Xu D, Liew FY. Protection against leishmaniasis by injection of DNA encoding a major surface glycoprotein, gp63, of L. major. Immunology. 1995;84(2):173–176.
  • Campos-Neto A, Webb JR, Greeson K, et al. 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(6):2828–2836.
  • Aguilar-Be I, da Silva Zardo R, Paraguai de Souza E, et al. Cross-protective efficacy of a prophylactic Leishmania donovani DNA vaccine against visceral and cutaneous murine leishmaniasis. Infect Immun. 2005;73(2):812–819.
  • Sukumaran B, Tewary P, Saxena S, et al. Vaccination with DNA encoding ORFF antigen confers protective immunity in mice infected with Leishmania donovani. Vaccine. 2003;21(11–12):1292–1299.
  • Borja-Cabrera GP, Santos FB, Picillo E, et al. Nucleoside hydrolase DNA vaccine against canine visceral leishmaniasis. Procedia Vaccinol. 2009;1(1):104–109.
  • Rafati S, Salmanian AH, Taheri T, et al. A protective cocktail vaccine against murine cutaneous leishmaniasis with DNA encoding cysteine proteinases of Leishmania major. Vaccine. 2001;19(25–26):3369–3375.
  • Iborra S, Soto M, Carrión J, et al. Vaccination with a plasmid DNA cocktail encoding the nucleosomal histones of Leishmania confers protection against murine cutaneous leishmaniosis. Vaccine. 2004;22(29–30):3865–3876.
  • Rodríguez-Cortés A, Ojeda A, López-Fuertes L, et al. Vaccination with plasmid DNA encoding KMPII, TRYP, LACK and GP63 does not protect dogs against Leishmania infantum experimental challenge. Vaccine. 2007;25(46):7962–7971.
  • Campbell K, Diao H, Ji J, et al. DNA immunization with the gene encoding P4 nuclease of Leishmania amazonensis protects mice against cutaneous Leishmaniasis. Infect Immun. 2003;71(11):6270–6278.
  • Xu D, McSorley SJ, Chatfield SN, et al. Protection against Leishmania major infection in genetically susceptible BALB/c mice by gp63 delivered orally in attenuated Salmonella typhimurium (AroA- AroD-). Immunology. 1995;85(1):1–7.
  • Walker PS, Scharton-Kersten T, Rowton ED, et al. Genetic immunization with glycoprotein 63 cDNA results in a helper T cell type 1 immune response and protection in a murine model of leishmaniasis. Hum Gene Ther. 1998;9(13):1899–1907.
  • Dumonteil E, Andrade-Narvarez F, Escobedo-Ortegon J, et al. Comparative study of DNA vaccines encoding various antigens against Leishmania mexicana. Dev Biol (Basel). 2000;104:135–141.
  • Dumonteil E, Maria Jesus RS, Javier EO. Maria del Rosario GM. DNA vaccines induce partial protection against Leishmania mexicana. Vaccine. 2003;21(17–18):2161–2168.
  • López-Fuertes L, Pérez-Jiménez E, Vila-Coro AJ, et al. DNA vaccination with linear minimalistic (MIDGE) vectors confers protection against Leishmania major infection in mice. Vaccine. 2002;21(3–4):247–257.
  • Gomes DC, Pinto EF, de Melo LD, et al. Intranasal delivery of naked DNA encoding the LACK antigen leads to protective immunity against visceral leishmaniasis in mice. Vaccine. 2007;25(12):2168–2172.
  • Marques-da-silva EA, Coelho EA, Gomes DC, et al. Intramuscular immunization with p36(LACK) DNA vaccine induces IFN-gamma production but does not protect BALB/c mice against Leishmania chagasi intravenous challenge. Parasitol Res. 2005;98(1):67–74.
  • Melby PC, Yang J, Zhao W, et al. Leishmania donovanip36(LACK) DNA vaccine is highly immunogenic but not protective against experimental Visceral Leishmaniasis. Infect Immun. 2001;69(8):4719–4725.
  • Kumar A, Samant M. DNA vaccine against visceral leishmaniasis: a promising approach for prevention and control. Parasite Immunol. 2016;38(5):273–281.
  • Gonzalo RM, Del Real G, Rodriguez JR, et al. A heterologous prime-boost regime using DNA and recombinant vaccinia virus expressing the Leishmania infantum P36/LACK antigen protects BALB/c mice from cutaneous leishmaniasis. Vaccine. 2002;20(7–8):1226–1231.
  • Kardani K, Bolhassani A, Shahbazi S. Prime-boost vaccine strategy against viral infections: mechanisms and benefits. Vaccine. 2016;34(4):413–423.
  • Ramiro MJ, Zárate JJ, Hanke T, et al. Protection in dogs against visceral leishmaniasis caused by Leishmania infantum is achieved by immunization with a heterologous prime-boost regime using DNA and vaccinia recombinant vectors expressing LACK. Vaccine. 2003;21(19–20):2474–2484.
  • Xiao-wen H, Shu-han S, Zhen-lin H, et al. Augmented humoral and cellular immune responses of a hepatitis B DNA vaccine encoding HBsAg by protein boosting. Vaccine. 2005;23(14):1649–1656.
  • Cui Z. DNA vaccine. Adv Genet. 2005;54:257–289.
  • Restifo NP, Ying H, Hwang L, et al. The promise of nucleic acid vaccines. Gene Ther. 2000;7(2):89–92.
  • Hobernik D, Bros M, Vaccines-How Far DNA. From Clinical Use? Int J Mol Sci. 2018;19(11):3605. doi: https://doi.org/10.3390/ijms19113605
  • Pandey SC, Kumar A, Samant M. Genetically modified live attenuated vaccine: a potential strategy to combat visceral leishmaniasis. Parasite Immunol. 2020;42(9):e12732.
  • Zabala-Peñafiel A, Todd D, Daneshvar H, et al. The potential of live attenuated vaccines against Cutaneous Leishmaniasis. Exp Parasitol. 2020;210:107849.
  • Titus RG, Gueiros-Filho FJ, de Freitas LA, et al. Development of a safe live Leishmania vaccine line by gene replacement. Proc Natl Acad Sci U S A. 1995;92(22):10267–10271.
  • Muyombwe A, Olivier M, Harvie P, et al. Protection against Leishmania major challenge infection in mice vaccinated with live recombinant parasites expressing a cytotoxic gene. J Infect Dis. 1998;177(1):188–195.
  • Kumari S, Samant M, Khare P, et al. Photodynamic vaccination of hamsters with inducible suicidal mutants of Leishmania amazonensis elicits immunity against visceral leishmaniasis. Eur J Immunol. 2009;39(1):178–191.
  • Davoudi N, Tate CA, Warburton C, et al. 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(9):1170–1177.
  • Mitchell GF, Handman E, Spithill TW. Vaccination against cutaneous leishmaniasis in mice using nonpathogenic cloned promastigotes of Leishmania major and importance of route of injection. Aust J Exp Biol Med Sci. 1984;62(Pt 2):145–153.
  • Gorczynski RM. Immunization of susceptible BALB/c mice against Leishmania braziliensis. II. Use of temperature-sensitive avirulent clones of parasite for vaccination purposes. Cell Immunol. 1985;94(1):11–20.
  • Rivier D, Shah R, Bovay P, et al. Vaccine development against cutaneous leishmaniasis. Subcutaneous administration of radioattenuated parasites protects CBA mice against virulent Leishmania major challenge. Parasite Immunol. 1993;15(2):75–84.
  • Kimsey PB, Theodos CM, Mitchen TK, et al. An avirulent lipophosphoglycan-deficient Leishmania major clone induces CD4+ T cells which protect susceptible BALB/c mice against infection with virulent L. major. Infect Immun. 1993;61(12):5205–5213.
  • Daneshvar H, Coombs GH, Hagan P, et al. Leishmania mexicana and Leishmania major: attenuation of wild-type parasites and vaccination with the attenuated lines. J Infect Dis. 2003;187(10):1662–1668.
  • Papadopoulou B, Roy G, Breton M, et al. Reduced infectivity of a Leishmania donovani biopterin transporter genetic mutant and its use as an attenuated strain for vaccination. Infect Immun. 2002;70(1):62–68.
  • Dey R, Meneses C, Salotra P, et al. Characterization of a Leishmania stage-specific mitochondrial membrane protein that enhances the activity of cytochrome c oxidase and its role in virulence. Mol Microbiol. 2010;77(2):399–414.
  • Dey R, Dagur PK, Selvapandiyan A, et al. Live attenuated Leishmania donovani p27 gene knockout parasites are nonpathogenic and elicit long-term protective immunity in BALB/c mice. J Immunol. 2013;190(5):2138–2149.
  • Carrión J, Folgueira C, Soto M, et al. Leishmania infantum HSP70-II null mutant as candidate vaccine against leishmaniasis: a preliminary evaluation. Parasit Vectors. 2011;4(1):150.
  • Solana JC, Ramírez L, Cook EC, et al. Subcutaneous Immunization of Leishmania HSP70-II null mutant line reduces the severity of the experimental Visceral Leishmaniasis in BALB/c mice. Vaccines (Basel). 2020;8(1).
  • Soto M, Ramírez L, Solana JC, et al. Inoculation of the Leishmania infantum HSP70-II null mutant induces long-Term protection against L. amazonensis Infection in BALB/c mice. Microorganisms. 2021;9(2):363. doi: https://doi.org/10.3390/microorganisms9020363.
  • Solana JC, Ramírez L, Corvo L, et al. Vaccination with a Leishmania infantum HSP70-II null mutant confers long-term protective immunity against Leishmania major infection in two mice models. PLoS Negl Trop Dis. 2017;11(5):e0005644.
  • Santi AMM, Lanza JS, Tunes LG, et al. Growth arrested live-attenuated Leishmania infantum KHARON1 null mutants display cytokinesis defect and protective immunity in mice. Sci Rep. 2018;8(1):11627.
  • Selvapandiyan A, Dey R, Nylen S, et al. Intracellular replication-deficient Leishmania donovani induces long lasting protective immunity against visceral leishmaniasis. J Immunol. 2009;183(3):1813–1820.
  • Karmakar S, Ismail N, Oliveira F, et al. Preclinical validation of a live attenuated dermotropic Leishmania vaccine against vector transmitted fatal visceral leishmaniasis. Commun Biol. 2021;4(1):1–14.
  • Zijlstra EE, el-Hassan AM, Ismael A, et al. Endemic kala-azar in eastern Sudan: a longitudinal study on the incidence of clinical and subclinical infection and post-kala-azar dermal leishmaniasis. Am J Trop Med Hyg. 994;51(6):826–836.
  • Bern C, Amann J, Haque R, et al. Loss of leishmanin skin test antigen sensitivity and potency in a longitudinal study of visceral leishmaniasis in Bangladesh. Am J Trop Med Hyg. 2006;75(4):744–748.
  • Montenegro J. 1926. A cutis-reação na leishmaniose. Anais da Faculdade de Medicina de São Paulo. 1:323–330.
  • Burza S, Croft SL, Boelaert M. Leishmaniasis. Lancet. 2018;392(10151):951–970.
  • Poulter LW, Seymour GJ, Duke O, et al. Immunohistological analysis of delayed-type hypersensitivity in man. Cell Immunol. 1982;74(2):358–369.
  • Cher DJ, Mosmann TR. Two types of murine helper T cell clone. II. Delayed-type hypersensitivity is mediated by TH1 clones. J Immunol. 1987;138(11):3688–3694.
  • Pacheco-Fernandez T, Volpedo G, Gannavaram S, et al. Revival of Leishmanization and Leishmanin. Front Cell Infect Microbiol. 2021;11:639801.
  • Mohebali M, Nadim A, Khamesipour A. An overview of leishmanization experience: a successful control measure and a tool to evaluate candidate vaccines. Acta Trop. 2019;200:105173.
  • Nadim A, Javadian E, Tahvildar-Bidruni G, et al. Effectiveness of leishmanization in the control of cutaneous leishmaniasis. Bull Soc Pathol Exot Filiales. 1983;76(4):377–383.
  • Nadim AJ, Mohebali E. The experience of leishmanization in the Islamic Republic of Iran. East Mediterr Health J. 1997;3(2): 284–289. M.
  • Fatemi M, Yaghoobi-Ershadi MR, Mohebali M, et al. The potential role of humans in the transmission cycle of leishmania major (Kinetoplastida: trypanosomatidae), the causative agent of the old World Zoonotic Cutaneous Leishmaniasis. J Med Entomol. 2018;55(6):1588–1593.
  • Denise H, Coombs GH, Mottram JC. Generation of Leishmania mutants lacking antibiotic resistance genes using a versatile hit-and-run targeting strategy. FEMS Microbiol Lett. 2004;235(1):89–94.
  • Seyed N, Peters NC, Rafati S. Translating observations from leishmanization into Non-Living Vaccines: the potential of dendritic cell-based vaccination strategies against. Front Immunol. 2018;9:1227.
  • Lestinova T, Rohousova I, Sima M, et al. Insights into the sand fly saliva: blood-feeding and immune interactions between sand flies, hosts, and Leishmania. PLoS Negl Trop Dis. 2017;11(7):e0005600.
  • Peters NC, Kimblin N, Secundino N, et al. Vector transmission of leishmania abrogates vaccine-induced protective immunity. PLoS Pathog. 2009;5(6):e1000484.
  • Peters NC, Bertholet S, Lawyer PG, et al. Evaluation of recombinant Leishmania polyprotein plus glucopyranosyl lipid A stable emulsion vaccines against sand fly-transmitted Leishmania major in C57BL/6 mice. J Immunol. 2012;189(10):4832–4841.
  • Stamper LW, Patrick RL, Fay MP, et al. Infection parameters in the sand fly vector that predict transmission of Leishmania major. PLoS Negl Trop Dis. 2011;5(8):e1288.
  • Halioua C. 2018OWJC: Why are drugs more profitable than vaccines? (Health economics working paper). (Ed.^(Eds) (Medium).
  • Dey R, Natarajan G, Bhattacharya P, et al. Characterization of cross-protection by genetically modified live-attenuated Leishmania donovani parasites against Leishmania mexicana. J Immunol. 2014;193(7):3513–3527.
  • Cooper MM, Loiseau C, McCarthy JS, et al. Human challenge models: tools to accelerate the development of malaria vaccines. Expert Rev Vaccines. 2019;18(3):241–251.
  • Ives A, Ronet C, Prevel F, et al. Leishmania RNA virus controls the severity of mucocutaneous leishmaniasis. Science. 2011;331(6018):775–778.