CC BY 4.0 · Journal of Child Science 2021; 11(01): e178-e184
DOI: 10.1055/s-0041-1731336
Review Article

Review of Development of Live Vaccines against Leishmaniasis

Mohammad Hossein Feiz Haddad
1   Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
2   Department of Parasitology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
,
Jalal Lomei
3   Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
,
Azar Shokri
4   Vector-borne Disease Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
,
Habib Habibpour
1   Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
2   Department of Parasitology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
,
Hossein Rezvan
5   Department of Pathobiology, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
,
Alireza Nourian
5   Department of Pathobiology, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
,
Mohammad Reza Mahmoudi
6   Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
› Author Affiliations
Funding This study was financially supported by Ahvaz Jundishapur University of Medical Sciences (grant no. CMRC-9820).
 

Abstract

Leishmaniasis is a serious public health problem in both tropical and temperate regions, caused by protozoan parasites of the genus Leishmania. Cutaneous leishmaniasis is the most common form of leishmaniasis worldwide. After recovery from the initial infection in most of the patients, a long-lasting natural immunity will be established. In individuals with HIV infection or in immune deficient patients, the more dangerous forms can occur. Despite many attempts, there is no efficient vaccine for leishmaniasis. The main concern for live-attenuated vaccines is the possibility of returning to the virulent form. Therefore, the safety is an important point in designing a successful vaccine. Nonvirulent parasites as vaccine candidates are achievable through gamma-irradiation, long-term culture, random mutations induced by chemical agents, and temperature-sensitive mutations. The type of change(s) in such parasites is not known well and drawbacks such as reversion to virulent forms was soon realized. Leishmania tarentolae with capacity of adaptation to mammalian system has a potential to be used as nonpathogenic vector in vaccine programs. Due to its nonpathogenic intrinsic property, it does not have the ability to replace with the pathogen form. Moreover, the main problems are associated with the production of live vaccines, including lyophilization, storage, standards, and quality control that must be considered. In this review, we focused on the importance of different approaches concerning the development of a live vaccine against leishmaniasis.


#

Introduction

Leishmaniasis, caused by an intracellular protozoan parasites of the genus Leishmania, is prevalent in subtropical and tropical regions all around the world.[1] [2] [3] The disease is considered as neglected tropical diseases with the incidence of 1.5 million new cases annually[4] [5] which leads to a considerable mortality and morbidity.[6] There are three main forms of the disease, namely, cutaneous leishmaniasis (CL), visceral leishmaniasis (VL), and mucocutaneous leishmaniasis (MCL).[7] VL, also known as kala-azar, is caused by Leishmania donovani (anthroponotic form) and L. infantum (zoonotic form). VL is the most important and severe form of leishmaniasis with 500,000 new cases annually. In this form of the disease, the parasites invade in internal organs such as liver and spleen. VL can lead to death in untreated cases and there is no effective vaccine for it.[8] MCL is mainly caused by L. braziliensis complex in mucosal tissues of nose and mouth and can result in massive tissue damage and permanent disfiguration.[9] CL is self-healing disease[10] caused mostly by the L. tropica and L. major in the Central Asia and Middle East[11] and L. braziliensis and L. mexicana complexes in the Americas.[12] CL is the most common form of leishmaniasis in the world, and 90% of all CL cases occur only in seven countries: Syria, Iran, Saudi Arabia, Algeria, Peru, Brazil, and Afghanistan.[1] CL can lead to skin deformities, and in immune compromised patients giving rise to the diffuse cutaneous leishmaniasis.[13] The host immune response has a crucial role in leishmaniasis, both in efficacy of treatment and the clinical patterns of the disease.[14] Success in development of vaccine depends on selecting an appropriate vaccine candidate and the immune biology of pathogen/host interactions.[15] Cell-mediated immunity particularly CD4 T cells are critical in leishmaniasis and protection against the disease.[16] The host defense mainly relies on response of Th1 cells which were activated through interleukin (IL)-12. Antigen presenting cells (APCs) and T cells, as they produce IL-12 and interferon gamma (IFN-γ), are crucial for limiting the numbers of leishmania parasites.[17] In contrast, Th2 cells produce cytokines, mainly IL-4, IL-5, and IL-13 and anti-inflammatory cytokines that have suppressive effect on host immunity which leads to parasite survival during leishmaniasis.[17] [18] These effects are exclusively studied in CL and murine models.[7] [19] Generation of proper immunologic memory is the main challenge in designing a vaccine that can cause effective immunity. Different studies revealed that long-lasting immunity against leishmaniasis needs to involve the function of both regulatory T cells (Tregs) and Th1-mediated immune response.[20] [21] Currently, treatment of any form of leishmaniasis is remarkably dependent on pentavalent antimonials as first-line and amphotericin B as second-line drugs.[22] This treatment is usually adequate but long duration of treatment, high costs, and side effects are still challenging. Reversion to virulence form has been reported in host with weak immune system, especially in HIV infected individuals.[23] Furthermore, strains which are resistant to available drugs created the need for an effective vaccine.[24] Currently, there is no impermissible vaccine against human leishmaniasis, even several vaccines have developed to clinical trials, most are still in early research phases and need more develop ([Table 1]).[25] [26] [27] From 2004, four vaccines have been licensed against canine leishmaniosis, two in Brazil (Leishmune, license was withdrawn in 2014, and Leish-Tec) and two in Europe (CaniLeish and LetiFend).[28] Efforts for finding an effective vaccine against leishmaniasis is continuing worldwide.[9] [29] Finally, development of an effective vaccine is one of the most cost-effective ways to eliminate and/or control CL and VL. This review aims to discuss the importance of different approaches concerning the development of a live-attenuated vaccine against leishmaniasis.

Table 1

Development status of current vaccine candidates

Candidate name/identifier

Preclinical

Phase I

Phase II

Reference

LEISH-F2

X

[11] [25]

LEISH-F3

X

[11] [26]

Various Lutzomyia and fly antigens

X

[11] [27]

Various second-generation protein-based vaccines

X

[11] [27]

Various third-generation DNA-based and heterologous prime-boost vaccines

X

[11] [27]


#

Live Active Vaccines

Leishmanization (LZ) was the first effort in using live parasite for human immunization, which was used in some countries for immunization against CL. There is a constant concern about revision to virulent type. This method was used in last century in several countries particularly in the Middle East, but it countered severe barriers such as local lesions in 2 to 3% of cases. The program was stopped due to some problems such as spreading the HIV infection, increasing the use of immunosuppressive drugs, permanent skin lesions, dissemination of lesions, ethical reasons, and problems in quality control of inoculant and long-lasting parasite persistence. Uzbekistan is one of the endemic countries, which LZ is licensed and its efficacy in human trials was proven. Efforts for improving the safety of this practice is continuing as this program can lead to an efficient vaccine against CL. It is declared that under controlled condition, it is achievable to elicit quick immune responses using the killed parasites and adjuvants.[30]


#

Live-Attenuated Vaccine

Several methods such as long-term in vitro cultures,[31] attenuation with temperature,[32] chemical mutagenesis,[33] [34] culture under drug pressure,[35] and γ-attenuation have been used to develop an attenuated Leishmania strains for many years. Recently, live-attenuated mutants through the gene alteration have revealed ability to induce proper immune responses in experimental hosts ([Table 2]). This helps researchers to identify genes which are necessary for Leishmania parasite's survival and/or for their virulence.[33] [49] [50] [51] [52] [53] [54]

Table 2

Defined and undefined genetic alteration live-attenuated vaccines against leishmaniasis

Species

Attenuation process

Model

Outcome of immunization

References

L. tropica, L. major

Long-term in vitro culture

BALB/c and C57BL/6

Persistent low-grade cutaneous disease, BALB/c: partially protection C57BL/6: completely resistant

[31]

L. major

Long-term in vitro culture

BALB/c

Protection

[36]

L. chagasi

Long-term in vitro culture

BALB/c

No protection

[37]

L. amazonensis

Long-term in vitro culture

C57BL/6

Decrease parasite load, increased IFN-γ, smaller lesions

[38]

L. braziliensis

Temperature sensitivity

BALB/c

Protection

[32]

L. major

Radio attenuated

CBA

Resistance to subsequent infection with L. mexicana

[39]

L. major

Gamma irradiation

BALB/c and CBA

Protection against heterologous and homologs challenge

[33]

L. infantum

In vitro under gentamicin pressure

Dog

In the endemic area against canine visceral leishmaniosis gentamicin-attenuated L. infantum induced a strong and significant protective effect

[40] [41] [42]

L. infantum

KHARON1 (KH1) null mutants (ΔLikh1)

BALB/c

Mice

ΔLikh1-immunized mice presented reduced parasite burden upon challenging with virulent L. infantum, when compared with naïve mice. An effect associated with increased IL-17 production and Li SLA-specific IgG serum levels

[43]

L. infantum

HSP70-II genes null mutant (LiΔHSP70-II)

BALB/c and C57BL/6

LiΔHSP70-II attenuated line activates mammalian immune system for inducing moderate proinflammatory responses

[44]

L. donovani

LdCen(−/−) centrin deleted

Mice and hamster

LdCen−/− live-attenuated vaccine induced a strong antibody production, a selectively CD4+ and CD8 + T cell activation in addition to prominent type 1 immune response that contributed to a remarkable reduction in bone marrow parasite load, even 24 months post-L. infantum infection

[45]

L. donovani

Arabino-1, 4-lactone oxidase (ΔALO) null mutants

BALB/c

Mice

Live-attenuated ΔALO parasites are induce protective immunity, safe, and can provide sustained protection against L. donovani

[46]

L. donovani

p27 gene-deleted (Ldp27−/−), Ldp27episomal add-back (Ldp27−/−AB), Centrin1 gene deleted (LdCen−/−), and LdCen episomal add-back (LdCen−/−AB) lines of L. donovani

BALB/c mice

Attenuated parasite-infected mice induced higher IL-2 and IFN-γ but significantly less IL-10 production by ovalbumin-specific CD4+ T cells, resulting in the proliferation of Th1 cells

[47]

L. donovani

Centrin1 and p27 genes deleted live-attenuated Leishmania parasites (LdCen1−/− and Ldp27−/−)

Human macrophages

LdCen1−/− and Ldp27−/− strongly stimulated production of proinflammatory cytokines including, IL-12, IFN-γ, TNF-α, IL-2, IL-6, and IL-17 in the PBMCs

[48]

Abbreviations: IFN-γ, interferon gamma; IgG, immunoglobulin G; IL, interleukin; TNF-α, tumor necrosis factor α.


In summary, it is believed that live-attenuated parasites can be considered suitable vaccine candidate as they present antigens to the APCs and activate them to induce immunity through mimicking natural infection and lead to the highest rate of CD4+ T cells polarization.[55] Also, it evaluates the memory of the immune system due to delivery of whole antigens instead of its subunits, and finally, they cause antigen constancy through long time subclinical infection. Therefore, memory cells and subsequently antigen-specific responses will be produced and react to reinfection.[56] A significant protection by attenuated parasites in murine models have been approved, but there is forever reversion capability that makes them inefficient for using them in human vaccination programs. There is a great risk about consequent reactivation in HIV/Leishmania coinfected individuals along with increasing the asymptomatic cases. Additionally, attenuation may lead to poor immune response as the strains are not able to induce subclinical infection or they are not able to express necessary antigenic epitopes. Despite of satisfactory experimental results, the important safety points are needed for attention before using genetically attenuated parasitic vaccines. For achieving an effective memory response against Leishmania parasites, persistence of antigen by producing subclinical infections is worthwhile. Reactivation of Leishmania is shown in immunocompromised patients. This proves the importance of careful investigation of live-attenuated parasites which can cause subclinical infection.[30]

Based on the used attenuation procedure, live-attenuated parasites are divided in two groups: defined and undefined genetically modified ([Table 2]).[57] In defined attenuation, by specific mutagenesis, the parasites lost the ability of encoding one or more essential virulence genes. Lazer irradiation or chemical mutagenesis or long-term in vitro culture can produce undefined attenuated parasites. In clinical trial performed in Iran with live vaccine, the researchers used stabilates which were made from stationary phase of parasites. They performed two trials in adults and followed them till healing the leishmanial lesions.[15] [58] There is a great challenge in using of live-attenuated Leishmania in the regions with high risk of HIV infection. Nowadays, the approved live-attenuated vaccines for clinical trials are made from L. major [59] and that means the vaccine will be prepared soon and will be able to help protect masses against the other coexistence species in endemic areas.[6]


#

Genetically Live-Attenuated Vaccines

Genetically modified vaccines have been developed in recent years and are powerful tools for creating immunity.[59] For achieving a protective vaccine, modifying some of Leishmania genes through deleting virulent or necessary genes seems to be successful.[21] The transfection technology through modifying one or more genes in Leishmania parasite leads to a great affect. The DNA fragments could be transferred through physical methods such as electroporation to the parasite's nuclei.[60] Antibiotic-resistant genes in a linear structure need to be combined with the genome through homologous recombination for removing a gene. Antibiotic-resistant genes in a linear form should be constructed and need to integrated by homolog recombining for removing a gene. This leads to transferring the sequence of DNA in genome of Leishmania parasite.[60] To achieve an absolute knockout, there should be another construct to produce antibiotic-resistant gene by Leishmania parasite to set the alleles of the second gene. This manipulation leads to parasites new phenotype and features which hereditary transfer to the next generation. The main concern in gene targeting is the new gene location control as it is important that it should not interfere with genes normal function. Although the genome of Leishmania parasite is easily manipulated, the site of entered gene is important and should be confirmed by molecular genetics assays. Another important issue after gene transfer is the phenotype alterations and need to be studied more. In this regard, one of the famous experiments was performed by Titus et al who decided to vaccinate mice against virulent L. major parasite[49] with dihydrofolate reductase-thymidylate synthase null mutant of the parasite, which they achieved through gene targeting. Unfortunately, it did not show persistent protection affect in susceptible mouse strains and in Rhesus monkeys[61] but revealed the resistant phenotype of L. major parasite.[49] Another attempt on mice and hamster, using mutants of manipulated L. Mexicana which lacked cysteine proteinase genes, showed promising results such as low parasite burden, smaller lesions, and delay in disease onset in animals were observed.[52] [62] These findings show that the protective immune response is achievable through genetic attenuation of live Leishmania parasite. In a study performed by Uzonna et al, they found that very susceptible mice were protected after vaccination with phosphoglycan (PG)-deficient L. major without considerable Th1 response.[54] Experiments with BT1 null mutant of L. donovani in infecting mice in comparison to wild type were promising and the susceptible individuals were able to attain protective immunity against the parasite.[53] Other researchers proved the efficacy of live genetically attenuated Leishmania vaccines in inducing immunity as Silvestre et al demonstrated the production of IFN-γ/ IL-10 in SIR2-deficient L. infantum that seems to be protective.[63] Other researchers tried to attain an effective vaccine with elimination of all PGs through deleting the lipophosphoglycan (LPG) gene in L. major. This leads to creating a nonpathogen parasite, which obtained life ability at low level for more than 2 years in mice. Thus, immunization with lpg2 parasites in very susceptible BALB/c mice induced protection against virulent L. major parasites.[12] In another research performed by Kedzierski et al, vaccination with a virulent L. major with absence of phosphomannomutase in susceptible BALB/c mice induced a protective response. They declared that increase in Th cells and their role in lymph nodes during the infection and low levels of IL-13 and IL-10 are the main reason of protection against Leishmania parasites.[64] In a 2009 study, it was revealed that by using of centrin gene (LdCen1−/−) disrupted L. donovani parasites were able to protect susceptible BALB/c mice and immune-deficient mice and hamsters against infection with L. braziliensis.[65] Fiuza et al showed that vaccination of dogs with LdCen−/− vaccine leads to Type 1 polarization and Type 2 inhibition along with production of antibody.[62] It is shown by Dey et al that knockout of protein 27 (Ldp27−/−) in amastigotes of L. donovani can induce protection against other Leishmania species.[47] Mutant of L. infantum which lacks both HSP70-II (DHSP70-II) alleles can protect BALB/c mice of infection with L. major, resulting in production of NO and type 1 immune responses with immunoglobulin G subclass analyses as reported by Carrion et al.[66] New generation of live-attenuated vaccine from L. major strain (LmCen−/−) for LZ has been explained by Zhang et al as they suggested effective protection against leishmaniasis without causing lesion. It seems the gene-free vaccine can be considered for human clinical trials in phase I.[7] Some limitations including concern about probability of reversion to virulent form and using them in immune-deficient people and manufacturing problems make the use of these vaccine more difficult.


#

Nonpathogenic Leishmania (Leishmania tarentolae)

In a study conducted by Breton et al, the use of nonpathogenic Leishmania strain (L. tarentolae) as a vector for designing a vaccine for VL is suggested.[67] The strain is harmless for human and in vitro evaluations revealed that the parasites are able to infect both human and murine phagocytic cells specially dendrite cells (DCs). Also, the parasites are able to promote DC maturation through monitoring their surface activation markers such as MHC-II, CD40, and CD83.[67] [68] The results obtained from experiments of L. tarentolae are comparable with the results of L. major infection in recruiting immune cells and producing inflammatory cytokines. Analysis of genome sequence has shown the similarity between pathogenic species including L. major, L. braziliensis, and L. infantum and 19% similarity between the shared genes in all species.

This suggests that some of these genes are related to the capacity of pathogenicity in this strain which makes it harmless to human. For instance, the presence of high copy number of amastin family in pathogenic species of Leishmania and presence of just two copies of L. tarentolae [69] and this is the reason of L. tarentolae's inability for application in mammalian macrophages. A protective immune response against L. donovani in susceptible BALB/c mice has been induced by experimental intraperitoneal administration of L. tarentolae that leads to a conclusion that this was caused through presentation of enhanced antigen and effective response of Th1 cells.[67] By considering the ability of gene manipulation and producing transgenic L. tarentolae which can be immune dominant antigens of Leishmania parasite, it would be considered as an effective vector in antileishmanial vaccines. The plan is using this strain of Leishmania as a special delivering and expressing leishmanial antigens system in host. Also, the A2 protein which is expressed by L. donovani complex and promotes visceralization of parasite is not present in L. tarentolae. Recent studies have demonstrated that the absence of virulence genes including LPG3, CPB, GP63, and amastin is not related to the lack of pathogenicity in L. tarentolae.[70] In a study, administration of A2-recombinant L. tarentolae vaccine was induced protection against L. infantum challenge in BALB/c mice and the protection was associated with production of high levels of IFN-γ before and after challenge.[71] As L. tarentolae is able to induce T-cells-mediated protection, it could be considered as a safe vector for live vaccines against Leishmania parasites and possibly other intracellular pathogens. Also, infection with L. tarentolae can elicit maturation of DC which are responsible for parasite phagosytose.[72] Salehi et al designed and evaluated a live vaccine by using recombinant E7 protein which was expressed by L. tarentolae for protection of mice against human papillomavirus–associated tumors. Interestingly, the vaccine showed the best protection against TC-1-induced tumors73. Comparative studies on L. tarentolae can be helpful for understanding the pathogenicity of Leishmania and its relationships with the host.


#

Conclusion

Despite the efforts of achieving an effective vaccine against leishmaniasis, there is still a long way to meet the vaccine development goals. Easy adaptation of Leishmania tarentolae and its safety to mammalian cells is promising to use it as a vector in live vaccines. However, it is important to take in considerations the major problems associated with production of live vaccines, including lyophilization, storage, standards, and quality control.


#
#

Conflict of Interest

None declared.

Note

The results described in this article were part of PhD student thesis of H.H.


  • References

  • 1 Torres-Guerrero E, Quintanilla-Cedillo MR, Ruiz-Esmenjaud J, Arenas R. Leishmaniasis: a review. F1000 Res 2017; 6: 750-750
  • 2 Feiz Haddad MH, Ghasemi E, Maraghi S, Tavala M. Identification of Leishmania species isolated from human cutaneous leishmaniasis in Mehran, Western Iran using nested PCR. Iran J Parasitol 2016; 11 (01) 65-72
  • 3 Hamoon NavardS, Rezvan H, Feiz Haddad MH. et al. Therapeutic effects of mesenchymal stem cells on cutaneous leishmaniasis lesions caused by Leishmania major . J Glob Antimicrob Resist 2020; 23: 243-250
  • 4 Hotez PJ, Alvarado M, Basáñez M-G. et al. The global burden of disease study 2010: interpretation and implications for the neglected tropical diseases. PLoS Negl Trop Dis 2014; 8 (07) e2865
  • 5 Vos T, Barber RM, Bell B. et al; Global Burden of Disease Study 2013 Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 2015; 386 (9995): 743-800
  • 6 Zabala-Peñafiel A, Todd D, Daneshvar H, Burchmore R. The potential of live attenuated vaccines against cutaneous leishmaniasis. Exp Parasitol 2020; 210: 107849
  • 7 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 (01) 3461
  • 8 Khatoon N, Pandey RK, Prajapati VK. Exploring Leishmania secretory proteins to design B and T cell multi-epitope subunit vaccine using immunoinformatics approach. Sci Rep 2017; 7 (01) 8285
  • 9 Mohammed ASA, Tian W, Zhang Y, Peng P, Wang F, Li T. Leishmania lipophosphoglycan components: a potent target for synthetic neoglycoproteins as a vaccine candidate for leishmaniasis. Carbohydr Polym 2020; 237: 116120
  • 10 Bennis I, De Brouwere V, Belrhiti Z, Sahibi H, Boelaert M. Psychosocial burden of localised cutaneous leishmaniasis: a scoping review. BMC Public Health 2018; 18 (01) 358
  • 11 Gillespie PM, Beaumier CM, Strych U, Hayward T, Hotez PJ, Bottazzi ME. Status of vaccine research and development of vaccines for leishmaniasis. Vaccine 2016; 34 (26) 2992-2995
  • 12 de Vasquez AM, Saenz RE, Petersen JL, Christensen HA, Johnson CM. Leishmania mexicana complex: human infections in the Republic of Panama. Am J Trop Med Hyg 1990; 43 (06) 619-622
  • 13 Desjeux P. Leishmaniasis: current situation and new perspectives. Comp Immunol Microbiol Infect Dis 2004; 27 (05) 305-318
  • 14 Khalili G, Dobakhti F, Mahmoudzadeh-Niknam H, Khaze V, Partovi F. Immunotherapy with imiquimod increases the efficacy of glucantime therapy of Leishmania major infection. Iran J Immunol 2011; 8 (01) 45-51
  • 15 Srivastava S, Shankar P, Mishra J, Singh S. Possibilities and challenges for developing a successful vaccine for leishmaniasis. Parasit Vectors 2016; 9 (01) 277
  • 16 Bogdan C. Mechanisms and consequences of persistence of intracellular pathogens: leishmaniasis as an example. Cell Microbiol 2008; 10 (06) 1221-1234
  • 17 Maspi N, Abdoli A, Ghaffarifar F. Pro- and anti-inflammatory cytokines in cutaneous leishmaniasis: a review. Pathog Glob Health 2016; 110 (06) 247-260
  • 18 Hurdayal R, Brombacher F. The role of IL-4 and IL-13 in cutaneous leishmaniasis. Immunol Lett 2014; 161 (02) 179-183
  • 19 Kaye P, Scott P. Leishmaniasis: complexity at the host-pathogen interface. Nat Rev Microbiol 2011; 9 (08) 604-615
  • 20 Belkaid Y, Piccirillo CA, Mendez S, Shevach EM, Sacks DL. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature 2002; 420 (6915): 502-507
  • 21 Mendez S, Reckling SK, Piccirillo CA, Sacks D, Belkaid Y. Role for CD4(+) CD25(+) regulatory T cells in reactivation of persistent leishmaniasis and control of concomitant immunity. J Exp Med 2004; 200 (02) 201-210
  • 22 Tiuman TS, Santos AO, Ueda-Nakamura T, Filho BP, Nakamura CV. Recent advances in leishmaniasis treatment. Int J Infect Dis 2011; 15 (08) e525-e532
  • 23 Bentwich Z. Concurrent infections that rise the HIV viral load. J HIV Ther 2003; 8 (03) 72-75
  • 24 Croft SL, Sundar S, Fairlamb AH. Drug resistance in leishmaniasis. Clin Microbiol Rev 2006; 19 (01) 111-126
  • 25 Duthie MS, Raman VS, Piazza FM, Reed SG. The development and clinical evaluation of second-generation leishmaniasis vaccines. Vaccine 2012; 30 (02) 134-141
  • 26 Nagill R, Kaur S. Vaccine candidates for leishmaniasis: a review. Int Immunopharmacol 2011; 11 (10) 1464-1488
  • 27 Rezvan H, Moafi M. An overview on Leishmania vaccines: a narrative review article. Vet Res Forum 2015; 6 (01) 1-7
  • 28 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 (05) 540-557
  • 29 Bezerra IPS, Costa-Souza BLS, Carneiro G, Ferreira LAM, de Matos Guedes HL, Rossi-Bergmann B. Nanoencapsulated retinoic acid as a safe tolerogenic adjuvant for intranasal vaccination against cutaneous leishmaniasis. Vaccine 2019; 37 (28) 3660-3667
  • 30 Saljoughian N, Taheri T, Rafati S. Live vaccination tactics: possible approaches for controlling visceral leishmaniasis. Front Immunol 2014; 5: 134
  • 31 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
  • 32 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 (01) 11-20
  • 33 Rivier D, Shah R, Bovay P, Mauel J. Vaccine development against cutaneous leishmaniasis. Subcutaneous administration of radioattenuated parasites protects CBA mice against virulent Leishmania major challenge. Parasite Immunol 1993; 15 (02) 75-84
  • 34 Kimsey PB, Theodos CM, Mitchen TK, Turco SJ, Titus RG. 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
  • 35 Daneshvar H, Coombs GH, Hagan P, Phillips RS. Leishmania mexicana and Leishmania major: attenuation of wild-type parasites and vaccination with the attenuated lines. J Infect Dis 2003; 187 (10) 1662-1668
  • 36 Segovia M, Artero JM, Mellado E, Chance ML. Effects of long-term in vitro cultivation on the virulence of cloned lines of Leishmania major promastigotes. Ann Trop Med Parasitol 1992; 86 (04) 347-354
  • 37 Streit JA, Recker TJ, Filho FG, Beverley SM, Wilson ME. Protective immunity against the protozoan Leishmania chagasi is induced by subclinical cutaneous infection with virulent but not avirulent organisms. J Immunol 2001; 166 (03) 1921-1929
  • 38 Gualdieri L, Rinaldi L, Petrullo L. et al. Intestinal parasites in immigrants in the city of Naples (southern Italy). Acta Trop 2011; 117 (03) 196-201
  • 39 Alexander J. A radioattenuated Leishmania major vaccine markedly increases the resistance of CBA mice to subsequent infection with Leishmania mexicana . Trans R Soc Trop Med Hyg 1982; 76 (05) 646-649
  • 40 Chettri JK, Deshmukh S, Holten-Andersen L, Jafaar RM, Dalsgaard I, Buchmann K. Comparative evaluation of administration methods for a vaccine protecting rainbow trout against Yersinia ruckeri O1 biotype 2 infections. Vet Immunol Immunopathol 2013; 154 (1-2): 42-47
  • 41 Daneshvar H, Molaei MM, Kamiabi H, Burchmore R, Hagan P, Stephen Phillips R. Gentamicin-attenuated Leishmania infantum: cellular immunity production and protection of dogs against experimental canine leishmaniasis. Parasite Immunol 2010; 32 (11-12): 722-730
  • 42 Daneshvar H, Sedghy F, Dabiri S. et al. Alteration in mononuclear cell subpopulations in dogs immunized with gentamicin-attenuated Leishmania infantum . Parasitology 2012; 139 (13) 1689-1696
  • 43 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 (01) 11627
  • 44 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 (05) e0005644
  • 45 Fiuza JA, Gannavaram S, Santiago HdaC. et al. Vaccination using live attenuated Leishmania donovani centrin deleted parasites induces protection in dogs against Leishmania infantum . Vaccine 2015; 33 (02) 280-288
  • 46 Anand S, Madhubala R. Genetically engineered ascorbic acid-deficient live mutants of Leishmania donovani induce long lasting protective immunity against visceral leishmaniasis. Sci Rep 2015; 5: 10706
  • 47 Dey R, Dagur PK, Selvapandiyan A, Mccoy JP, Salotra P, Duncan R. 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: 2138-2149 DOI: 10.4049/jimmunol.1202801a.
  • 48 Avishek K, Kaushal H, Gannavaram S. et al. Gene deleted live attenuated Leishmania vaccine candidates against visceral leishmaniasis elicit pro-inflammatory cytokines response in human PBMCs. Sci Rep 2016; 6: 33059
  • 49 Titus RG, Gueiros-Filho FJ, de Freitas LA, Beverley SM. Development of a safe live Leishmania vaccine line by gene replacement. Proc Natl Acad Sci U S A 1995; 92 (22) 10267-10271
  • 50 Dumas C, Ouellette M, Tovar J. et al. Disruption of the trypanothione reductase gene of Leishmania decreases its ability to survive oxidative stress in macrophages. EMBO J 1997; 16 (10) 2590-2598
  • 51 Hübel A, Krobitsch S, Hörauf A, Clos J. Leishmania major Hsp100 is required chiefly in the mammalian stage of the parasite. Mol Cell Biol 1997; 17 (10) 5987-5995
  • 52 Alexander J, Coombs GH, Mottram JC. Leishmania mexicana cysteine proteinase-deficient mutants have attenuated virulence for mice and potentiate a Th1 response. J Immunol 1998; 161 (12) 6794-6801
  • 53 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 (01) 62-68
  • 54 Uzonna JE, Späth GF, Beverley SM, Scott P. Vaccination with phosphoglycan-deficient Leishmania major protects highly susceptible mice from virulent challenge without inducing a strong Th1 response. J Immunol 2004; 172 (06) 3793-3797
  • 55 Spörri R, Reis e Sousa C. Inflammatory mediators are insufficient for full dendritic cell activation and promote expansion of CD4+ T cell populations lacking helper function. Nat Immunol 2005; 6 (02) 163-170
  • 56 Foulds KE, Wu CY, Seder RA. Th1 memory: implications for vaccine development. Immunol Rev 2006; 211 (01) 58-66
  • 57 Silvestre R, Cordeiro-da-Silva A, Ouaissi A. Live attenuated Leishmania vaccines: a potential strategic alternative. Arch Immunol Ther Exp (Warsz) 2008; 56 (02) 123-126
  • 58 Khamesipour A, Rafati S, Davoudi N, Maboudi F, Modabber F. Leishmaniasis vaccine candidates for development: a global overview. Indian J Med Res 2006; 123 (03) 423-438
  • 59 Alvar J, Croft SL, Kaye P, Khamesipour A, Sundar S, Reed SG. Case study for a vaccine against leishmaniasis. Vaccine 2013; 31 (Suppl. 02) B244-B249
  • 60 Papadopoulou B, Dumas C. Parameters controlling the rate of gene targeting frequency in the protozoan parasite Leishmania . Nucleic Acids Res 1997; 25 (21) 4278-4286
  • 61 Amaral VF, Teva A, Oliveira-Neto MP. et al. Study of the safety, immunogenicity and efficacy of attenuated and killed Leishmania (Leishmania) major vaccines in a rhesus monkey (Macaca mulatta) model of the human disease. Mem Inst Oswaldo Cruz 2002; 97 (07) 1041-1048
  • 62 Pulcini C, Massin S, Launay O, Verger P. Factors associated with vaccination for hepatitis B, pertussis, seasonal and pandemic influenza among French general practitioners: a 2010 survey. Vaccine 2013; 31 (37) 3943-3949
  • 63 Silvestre R, Cordeiro-Da-Silva A, Santarém N, Vergnes B, Sereno D, Ouaissi A. SIR2-deficient Leishmania infantum induces a defined IFN-γ/IL-10 pattern that correlates with protection. J Immunol 2007; 179 (05) 3161-3170
  • 64 Kedzierski L, Curtis JM, Doherty PC, Handman E, Kedzierska K. Decreased IL-10 and IL-13 production and increased CD44hi T cell recruitment contribute to Leishmania major immunity induced by non-persistent parasites. Eur J Immunol 2008; 38 (11) 3090-3100
  • 65 Selvapandiyan A, Dey R, Nylen S, Duncan R, Sacks D, Nakhasi HL. Intracellular replication-deficient Leishmania donovani induces long lasting protective immunity against visceral leishmaniasis. J Immunol 2009; 183 (03) 1813-1820
  • 66 Carrion J, Folgueira C, Soto M, Fresno M, Requena JM. Leishmania infantum HSP70-II null mutant as candidate vaccine against leishmaniasis: a preliminary evaluation. Parasit Vectors 2011; 4: 150 DOI: 10.1186/1756-3305-4-150.
  • 67 Breton M, Tremblay MJ, Ouellette M, Papadopoulou B. Live nonpathogenic parasitic vector as a candidate vaccine against visceral leishmaniasis. Infect Immun 2005; 73 (10) 6372-6382
  • 68 Taylor VM, Muñoz DL, Cedeño DL, Vélez ID, Jones MA, Robledo SM. Leishmania tarentolae: utility as an in vitro model for screening of antileishmanial agents. Exp Parasitol 2010; 126 (04) 471-475
  • 69 Raymond F, Boisvert S, Roy G. et al. Genome sequencing of the lizard parasite Leishmania tarentolae reveals loss of genes associated to the intracellular stage of human pathogenic species. Nucleic Acids Res 2012; 40 (03) 1131-1147
  • 70 Azizi H, Hassani K, Taslimi Y, Najafabadi HS, Papadopoulou B, Rafati S. Searching for virulence factors in the non-pathogenic parasite to humans Leishmania tarentolae . Parasitology 2009; 136 (07) 723-735
  • 71 Mizbani A, Taheri T, Zahedifard F. et al. Recombinant Leishmania tarentolae expressing the A2 virulence gene as a novel candidate vaccine against visceral leishmaniasis. Vaccine 2009; 28 (01) 53-62
  • 72 Breton M, Zhao C, Ouellette M, Tremblay MJ, Papadopoulou B. A recombinant non-pathogenic Leishmania vaccine expressing human immunodeficiency virus 1 (HIV-1) Gag elicits cell-mediated immunity in mice and decreases HIV-1 replication in human tonsillar tissue following exposure to HIV-1 infection. J Gen Virol 2007; 88 (Pt 1): 217-225
  • 73 Salehi M, Taheri T, Mohit E, Zahedifard F, Seyed N, Taslimi Y. et al. Recombinant Leishmania tarentolae encoding the HPV type 16 E7 gene in tumor mice model. Immunotherapy 2012; 4: 1107-1120 DOI: 10.2217/imt.12.110.

Address for correspondence

Habib Habibpour, PhD
Department of Parasitology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences
Ahvaz
Iran   

Publication History

Received: 29 August 2020

Accepted: 14 May 2021

Article published online:
26 June 2021

© 2021. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Torres-Guerrero E, Quintanilla-Cedillo MR, Ruiz-Esmenjaud J, Arenas R. Leishmaniasis: a review. F1000 Res 2017; 6: 750-750
  • 2 Feiz Haddad MH, Ghasemi E, Maraghi S, Tavala M. Identification of Leishmania species isolated from human cutaneous leishmaniasis in Mehran, Western Iran using nested PCR. Iran J Parasitol 2016; 11 (01) 65-72
  • 3 Hamoon NavardS, Rezvan H, Feiz Haddad MH. et al. Therapeutic effects of mesenchymal stem cells on cutaneous leishmaniasis lesions caused by Leishmania major . J Glob Antimicrob Resist 2020; 23: 243-250
  • 4 Hotez PJ, Alvarado M, Basáñez M-G. et al. The global burden of disease study 2010: interpretation and implications for the neglected tropical diseases. PLoS Negl Trop Dis 2014; 8 (07) e2865
  • 5 Vos T, Barber RM, Bell B. et al; Global Burden of Disease Study 2013 Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 2015; 386 (9995): 743-800
  • 6 Zabala-Peñafiel A, Todd D, Daneshvar H, Burchmore R. The potential of live attenuated vaccines against cutaneous leishmaniasis. Exp Parasitol 2020; 210: 107849
  • 7 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 (01) 3461
  • 8 Khatoon N, Pandey RK, Prajapati VK. Exploring Leishmania secretory proteins to design B and T cell multi-epitope subunit vaccine using immunoinformatics approach. Sci Rep 2017; 7 (01) 8285
  • 9 Mohammed ASA, Tian W, Zhang Y, Peng P, Wang F, Li T. Leishmania lipophosphoglycan components: a potent target for synthetic neoglycoproteins as a vaccine candidate for leishmaniasis. Carbohydr Polym 2020; 237: 116120
  • 10 Bennis I, De Brouwere V, Belrhiti Z, Sahibi H, Boelaert M. Psychosocial burden of localised cutaneous leishmaniasis: a scoping review. BMC Public Health 2018; 18 (01) 358
  • 11 Gillespie PM, Beaumier CM, Strych U, Hayward T, Hotez PJ, Bottazzi ME. Status of vaccine research and development of vaccines for leishmaniasis. Vaccine 2016; 34 (26) 2992-2995
  • 12 de Vasquez AM, Saenz RE, Petersen JL, Christensen HA, Johnson CM. Leishmania mexicana complex: human infections in the Republic of Panama. Am J Trop Med Hyg 1990; 43 (06) 619-622
  • 13 Desjeux P. Leishmaniasis: current situation and new perspectives. Comp Immunol Microbiol Infect Dis 2004; 27 (05) 305-318
  • 14 Khalili G, Dobakhti F, Mahmoudzadeh-Niknam H, Khaze V, Partovi F. Immunotherapy with imiquimod increases the efficacy of glucantime therapy of Leishmania major infection. Iran J Immunol 2011; 8 (01) 45-51
  • 15 Srivastava S, Shankar P, Mishra J, Singh S. Possibilities and challenges for developing a successful vaccine for leishmaniasis. Parasit Vectors 2016; 9 (01) 277
  • 16 Bogdan C. Mechanisms and consequences of persistence of intracellular pathogens: leishmaniasis as an example. Cell Microbiol 2008; 10 (06) 1221-1234
  • 17 Maspi N, Abdoli A, Ghaffarifar F. Pro- and anti-inflammatory cytokines in cutaneous leishmaniasis: a review. Pathog Glob Health 2016; 110 (06) 247-260
  • 18 Hurdayal R, Brombacher F. The role of IL-4 and IL-13 in cutaneous leishmaniasis. Immunol Lett 2014; 161 (02) 179-183
  • 19 Kaye P, Scott P. Leishmaniasis: complexity at the host-pathogen interface. Nat Rev Microbiol 2011; 9 (08) 604-615
  • 20 Belkaid Y, Piccirillo CA, Mendez S, Shevach EM, Sacks DL. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature 2002; 420 (6915): 502-507
  • 21 Mendez S, Reckling SK, Piccirillo CA, Sacks D, Belkaid Y. Role for CD4(+) CD25(+) regulatory T cells in reactivation of persistent leishmaniasis and control of concomitant immunity. J Exp Med 2004; 200 (02) 201-210
  • 22 Tiuman TS, Santos AO, Ueda-Nakamura T, Filho BP, Nakamura CV. Recent advances in leishmaniasis treatment. Int J Infect Dis 2011; 15 (08) e525-e532
  • 23 Bentwich Z. Concurrent infections that rise the HIV viral load. J HIV Ther 2003; 8 (03) 72-75
  • 24 Croft SL, Sundar S, Fairlamb AH. Drug resistance in leishmaniasis. Clin Microbiol Rev 2006; 19 (01) 111-126
  • 25 Duthie MS, Raman VS, Piazza FM, Reed SG. The development and clinical evaluation of second-generation leishmaniasis vaccines. Vaccine 2012; 30 (02) 134-141
  • 26 Nagill R, Kaur S. Vaccine candidates for leishmaniasis: a review. Int Immunopharmacol 2011; 11 (10) 1464-1488
  • 27 Rezvan H, Moafi M. An overview on Leishmania vaccines: a narrative review article. Vet Res Forum 2015; 6 (01) 1-7
  • 28 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 (05) 540-557
  • 29 Bezerra IPS, Costa-Souza BLS, Carneiro G, Ferreira LAM, de Matos Guedes HL, Rossi-Bergmann B. Nanoencapsulated retinoic acid as a safe tolerogenic adjuvant for intranasal vaccination against cutaneous leishmaniasis. Vaccine 2019; 37 (28) 3660-3667
  • 30 Saljoughian N, Taheri T, Rafati S. Live vaccination tactics: possible approaches for controlling visceral leishmaniasis. Front Immunol 2014; 5: 134
  • 31 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
  • 32 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 (01) 11-20
  • 33 Rivier D, Shah R, Bovay P, Mauel J. Vaccine development against cutaneous leishmaniasis. Subcutaneous administration of radioattenuated parasites protects CBA mice against virulent Leishmania major challenge. Parasite Immunol 1993; 15 (02) 75-84
  • 34 Kimsey PB, Theodos CM, Mitchen TK, Turco SJ, Titus RG. 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
  • 35 Daneshvar H, Coombs GH, Hagan P, Phillips RS. Leishmania mexicana and Leishmania major: attenuation of wild-type parasites and vaccination with the attenuated lines. J Infect Dis 2003; 187 (10) 1662-1668
  • 36 Segovia M, Artero JM, Mellado E, Chance ML. Effects of long-term in vitro cultivation on the virulence of cloned lines of Leishmania major promastigotes. Ann Trop Med Parasitol 1992; 86 (04) 347-354
  • 37 Streit JA, Recker TJ, Filho FG, Beverley SM, Wilson ME. Protective immunity against the protozoan Leishmania chagasi is induced by subclinical cutaneous infection with virulent but not avirulent organisms. J Immunol 2001; 166 (03) 1921-1929
  • 38 Gualdieri L, Rinaldi L, Petrullo L. et al. Intestinal parasites in immigrants in the city of Naples (southern Italy). Acta Trop 2011; 117 (03) 196-201
  • 39 Alexander J. A radioattenuated Leishmania major vaccine markedly increases the resistance of CBA mice to subsequent infection with Leishmania mexicana . Trans R Soc Trop Med Hyg 1982; 76 (05) 646-649
  • 40 Chettri JK, Deshmukh S, Holten-Andersen L, Jafaar RM, Dalsgaard I, Buchmann K. Comparative evaluation of administration methods for a vaccine protecting rainbow trout against Yersinia ruckeri O1 biotype 2 infections. Vet Immunol Immunopathol 2013; 154 (1-2): 42-47
  • 41 Daneshvar H, Molaei MM, Kamiabi H, Burchmore R, Hagan P, Stephen Phillips R. Gentamicin-attenuated Leishmania infantum: cellular immunity production and protection of dogs against experimental canine leishmaniasis. Parasite Immunol 2010; 32 (11-12): 722-730
  • 42 Daneshvar H, Sedghy F, Dabiri S. et al. Alteration in mononuclear cell subpopulations in dogs immunized with gentamicin-attenuated Leishmania infantum . Parasitology 2012; 139 (13) 1689-1696
  • 43 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 (01) 11627
  • 44 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 (05) e0005644
  • 45 Fiuza JA, Gannavaram S, Santiago HdaC. et al. Vaccination using live attenuated Leishmania donovani centrin deleted parasites induces protection in dogs against Leishmania infantum . Vaccine 2015; 33 (02) 280-288
  • 46 Anand S, Madhubala R. Genetically engineered ascorbic acid-deficient live mutants of Leishmania donovani induce long lasting protective immunity against visceral leishmaniasis. Sci Rep 2015; 5: 10706
  • 47 Dey R, Dagur PK, Selvapandiyan A, Mccoy JP, Salotra P, Duncan R. 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: 2138-2149 DOI: 10.4049/jimmunol.1202801a.
  • 48 Avishek K, Kaushal H, Gannavaram S. et al. Gene deleted live attenuated Leishmania vaccine candidates against visceral leishmaniasis elicit pro-inflammatory cytokines response in human PBMCs. Sci Rep 2016; 6: 33059
  • 49 Titus RG, Gueiros-Filho FJ, de Freitas LA, Beverley SM. Development of a safe live Leishmania vaccine line by gene replacement. Proc Natl Acad Sci U S A 1995; 92 (22) 10267-10271
  • 50 Dumas C, Ouellette M, Tovar J. et al. Disruption of the trypanothione reductase gene of Leishmania decreases its ability to survive oxidative stress in macrophages. EMBO J 1997; 16 (10) 2590-2598
  • 51 Hübel A, Krobitsch S, Hörauf A, Clos J. Leishmania major Hsp100 is required chiefly in the mammalian stage of the parasite. Mol Cell Biol 1997; 17 (10) 5987-5995
  • 52 Alexander J, Coombs GH, Mottram JC. Leishmania mexicana cysteine proteinase-deficient mutants have attenuated virulence for mice and potentiate a Th1 response. J Immunol 1998; 161 (12) 6794-6801
  • 53 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 (01) 62-68
  • 54 Uzonna JE, Späth GF, Beverley SM, Scott P. Vaccination with phosphoglycan-deficient Leishmania major protects highly susceptible mice from virulent challenge without inducing a strong Th1 response. J Immunol 2004; 172 (06) 3793-3797
  • 55 Spörri R, Reis e Sousa C. Inflammatory mediators are insufficient for full dendritic cell activation and promote expansion of CD4+ T cell populations lacking helper function. Nat Immunol 2005; 6 (02) 163-170
  • 56 Foulds KE, Wu CY, Seder RA. Th1 memory: implications for vaccine development. Immunol Rev 2006; 211 (01) 58-66
  • 57 Silvestre R, Cordeiro-da-Silva A, Ouaissi A. Live attenuated Leishmania vaccines: a potential strategic alternative. Arch Immunol Ther Exp (Warsz) 2008; 56 (02) 123-126
  • 58 Khamesipour A, Rafati S, Davoudi N, Maboudi F, Modabber F. Leishmaniasis vaccine candidates for development: a global overview. Indian J Med Res 2006; 123 (03) 423-438
  • 59 Alvar J, Croft SL, Kaye P, Khamesipour A, Sundar S, Reed SG. Case study for a vaccine against leishmaniasis. Vaccine 2013; 31 (Suppl. 02) B244-B249
  • 60 Papadopoulou B, Dumas C. Parameters controlling the rate of gene targeting frequency in the protozoan parasite Leishmania . Nucleic Acids Res 1997; 25 (21) 4278-4286
  • 61 Amaral VF, Teva A, Oliveira-Neto MP. et al. Study of the safety, immunogenicity and efficacy of attenuated and killed Leishmania (Leishmania) major vaccines in a rhesus monkey (Macaca mulatta) model of the human disease. Mem Inst Oswaldo Cruz 2002; 97 (07) 1041-1048
  • 62 Pulcini C, Massin S, Launay O, Verger P. Factors associated with vaccination for hepatitis B, pertussis, seasonal and pandemic influenza among French general practitioners: a 2010 survey. Vaccine 2013; 31 (37) 3943-3949
  • 63 Silvestre R, Cordeiro-Da-Silva A, Santarém N, Vergnes B, Sereno D, Ouaissi A. SIR2-deficient Leishmania infantum induces a defined IFN-γ/IL-10 pattern that correlates with protection. J Immunol 2007; 179 (05) 3161-3170
  • 64 Kedzierski L, Curtis JM, Doherty PC, Handman E, Kedzierska K. Decreased IL-10 and IL-13 production and increased CD44hi T cell recruitment contribute to Leishmania major immunity induced by non-persistent parasites. Eur J Immunol 2008; 38 (11) 3090-3100
  • 65 Selvapandiyan A, Dey R, Nylen S, Duncan R, Sacks D, Nakhasi HL. Intracellular replication-deficient Leishmania donovani induces long lasting protective immunity against visceral leishmaniasis. J Immunol 2009; 183 (03) 1813-1820
  • 66 Carrion J, Folgueira C, Soto M, Fresno M, Requena JM. Leishmania infantum HSP70-II null mutant as candidate vaccine against leishmaniasis: a preliminary evaluation. Parasit Vectors 2011; 4: 150 DOI: 10.1186/1756-3305-4-150.
  • 67 Breton M, Tremblay MJ, Ouellette M, Papadopoulou B. Live nonpathogenic parasitic vector as a candidate vaccine against visceral leishmaniasis. Infect Immun 2005; 73 (10) 6372-6382
  • 68 Taylor VM, Muñoz DL, Cedeño DL, Vélez ID, Jones MA, Robledo SM. Leishmania tarentolae: utility as an in vitro model for screening of antileishmanial agents. Exp Parasitol 2010; 126 (04) 471-475
  • 69 Raymond F, Boisvert S, Roy G. et al. Genome sequencing of the lizard parasite Leishmania tarentolae reveals loss of genes associated to the intracellular stage of human pathogenic species. Nucleic Acids Res 2012; 40 (03) 1131-1147
  • 70 Azizi H, Hassani K, Taslimi Y, Najafabadi HS, Papadopoulou B, Rafati S. Searching for virulence factors in the non-pathogenic parasite to humans Leishmania tarentolae . Parasitology 2009; 136 (07) 723-735
  • 71 Mizbani A, Taheri T, Zahedifard F. et al. Recombinant Leishmania tarentolae expressing the A2 virulence gene as a novel candidate vaccine against visceral leishmaniasis. Vaccine 2009; 28 (01) 53-62
  • 72 Breton M, Zhao C, Ouellette M, Tremblay MJ, Papadopoulou B. A recombinant non-pathogenic Leishmania vaccine expressing human immunodeficiency virus 1 (HIV-1) Gag elicits cell-mediated immunity in mice and decreases HIV-1 replication in human tonsillar tissue following exposure to HIV-1 infection. J Gen Virol 2007; 88 (Pt 1): 217-225
  • 73 Salehi M, Taheri T, Mohit E, Zahedifard F, Seyed N, Taslimi Y. et al. Recombinant Leishmania tarentolae encoding the HPV type 16 E7 gene in tumor mice model. Immunotherapy 2012; 4: 1107-1120 DOI: 10.2217/imt.12.110.