7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Building Research and Development Capacity for Neglected Tropical Diseases Impacting Leishmaniasis in the Middle East and North Africa: A Case Study

      editorial

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          There is an urgent need to build Research and Development (R&D) capacity for leishmaniasis and other neglected tropical diseases (NTDs) in the Middle East and North Africa. The neglected tropical diseases are a group of 17 viral, bacterial, protozoan, and helminthic infections that often have high morbidity but low mortality, causing disabilities through their impact on child development and education, pregnancy outcomes, and worker productivity [1]. NTDs promote poverty and interfere with economic development. Emerging information indicates that the Middle East and North Africa (MENA) region is highly endemic for several NTDs, with increasing levels of poverty and disease in the region [2]. Today, with the exception of Buruli ulcer, Chagas disease, dracunculiasis, and human African trypanosomiasis, the 13 remaining NTDs are found in MENA countries, with the most prevalent (both on and off the WHO’s list) being dengue virus, rabies, brucellosis, leprosy, trachoma, toxoplasmosis, cutaneous and visceral leishmaniasis, toxoplasmosis, facioliasis, schistosomiasis, and soil-transmitted helminth infections [2,3]. Nations such as Algeria, Iran, Libya, Morocco, Syria, Egypt, and Yemen are impoverished countries with especially high rates of NTDs [4]. Among all, Leishmania is the most important protozoan infection in the MENA region [5]. Rapid urbanization and human migration in this region has led to the spread of the sand fly vector and, consequently, leishmaniasis. Despite establishing national programs for sand fly vector control and leishmaniasis treatment, the disease continues to spread. Almost two-thirds of cutaneous leishmaniasis (CL) cases are reported from Afghanistan, Algeria, Brazil, Colombia, Iran, Syria, and Iraq [6]. Among the different countries of the MENA region, Syria is known to have the highest prevalence of CL (nearly 52,983 cases were reported in 2012). Incidence of visceral leishmaniasis (VL) in MENA is sporadic and has been reported mostly from Iraq (nearly 1,041 cases were reported in 2008), where 90% of cases occur in children under five years of age [6]. Globally, compared to HIV/AIDS, malaria, and tuberculosis, which receive 42.1% of the funds allocated to overseas development assistance for health, NTDs have generally been ignored, receiving only 0.6% of the budget [7]. Studies have also established that financial support for Research and Development for NTDs significantly lags behind AIDS and other better-known diseases [8]. While global efforts to eliminate some NTDs, such as lymphatic filariasis (http://www.afro.who.int/), have been successful in MENA countries, many have not received such attention. A similar level of neglect for both mass treatment of and R&D for NTDs has been noted for MENA countries. The World Health Organization (WHO) reports that mass drug administration coverage for many NTDs in MENA is low. For instance, the coverage for soil-transmitted helminthic infections in the WHO Eastern Mediterranean Region is less than one-half the coverage in sub-Saharan Africa [9]. In addition to mass treatments, we need to improve diagnosis and control of NTDs in MENA countries. This can be achieved by promoting R&D of new tools, including diagnostics and vaccines, and addressing challenges facing their implementation. So, how can we share critical information and identify the major obstacles to translating scientific breakthroughs into innovative strategies for reducing the burden of NTDs in MENA countries? We can achieve this goal by building research capacity that will promote support for the translation of laboratory findings into field applications, a crucial component for controlling the NTDs in the MENA region. Additionally, intervention and implementation research should focus on resolving barriers particular to MENA countries and translate into proper approaches for effective delivery of health interventions aimed at control or elimination of the targeted diseases. For example, a rapid response to outbreaks is generally poor in endemic MENA countries due to the absence of a good infrastructure for surveillance as well as lack of funding. In some cases, this is compounded by conflict and mass migrations that compromise the control of disease outbreaks in their early stages. In summary, two major tasks need to be undertaken to improve the control of NTDs in MENA: (i) increase research capacity in endemic MENA countries and (ii) translate laboratory discoveries into field applications appropriate for endemic MENA countries. As mentioned previously, leishmaniasis represents a good case study of the challenges facing control of NTDs in MENA. Today, MENA countries exhibit some of the highest incidences of cutaneous leishmaniasis globally [4], with worrisome increases in the war-torn areas of Syria, Iraq, and elsewhere. Besides the long-standing and crucial support of WHO-TDR (the Special Programme for Research and Training in Tropical Diseases), aimed at fostering an effective global research effort to control infectious diseases and promoting the translation of innovation to impact health in the MENA region, there are few other scientific activities that promoted leishmaniasis R&D in MENA countries during the past few years. A research and policy conference, LEISHMANIA: Collaborative Research Opportunities in North Africa and the Middle East, was held in June 2009 in Tunisia to promote international collaboration between the United States and the MENA countries most affected by leishmaniasis [10]. The conference was supported by the US National Institute of Allergy and Infectious Diseases Office of Global Research (OGR) and the National Institutes of Health (NIH) and hosted locally by the Institute Pasteur de Tunis. The developed collaborations and the outcome of this conference resulted in several successful grant proposals funded by the Civilian Research and Development Foundation from the OGR [11,12]. This remains only one successful example focused on leishmaniasis and there is a need for more of such supportive conferences to promote NTD research, especially in the MENA region. In July 2014, a European and Developing Countries Clinical Trials Partnership (EDCTP) forum on capacity development in Africa was held in Berlin. EDCTP was created in 2003 as a European response to the global health crises caused by three poverty-related diseases, including HIV/AIDS, tuberculosis, and malaria. The objective of the meeting was to identify current and emerging capacity development gaps in order to inform the development of the strategy and operational plans to control poverty-related diseases [13]. Similar to EDCTP, identifying the capacity development gaps in the MENA region, especially for highly endemic diseases such as cutaneous leishmaniasis, is highly recommended. Furthermore, creating a harmonized regional network to share information and experiences on different aspects of leishmaniasis is essential. These types of activities will help and promote opportunities to advise health authorities about the most effective measures for prevention and control of leishmaniasis, one of the highly prevalent diseases in MENA countries. In addition to conferences, by establishing regional R&D and training centers of excellence in the MENA region, we could promote and build long-lasting research skills and capacities and enhance the contribution of research to the control and elimination of NTDs. One such center was suggested recently for southern Europe [14], but a parallel center needs to be created in the MENA region. Such a training center could act as a resource for regional training activities that provide the building blocks for doing solid research on NTDs. It could also improve the ability of young researchers to establish a global network that promotes collaboration between institutions of the MENA region. By establishing a regional training center, sustainable capacity could be built through exchanging experiences and innovative approaches. Such a center would also foster good practices in health research to be commonly implemented by the researchers in this region. Iran is one of the MENA countries with the highest prevalence of zoonotic cutaneous leishmaniasis (Leishmania major), and the second-highest prevalence of anthroponotic cutaneous leishmaniasis (Leishmania tropica infection) [4]. With more than a decade of support from WHO-TDR, Iran led the development of first-generation leishmanization activities, which provide the underpinnings for more advanced vaccine development [15]. There are different universities and institutes in Iran with excellent scientific infrastructures that could act as a regional training center for leishmaniasis and other NTDs in the MENA region. Among all, Pasteur Institute of Iran is the oldest; it was established in 1920 in Tehran, using the Pasteur Institute of Paris as a template. Pasteur Institute of Iran has a close and active cooperation with international organizations such as the WHO, WHO-TDR, United Nations Children’s Fund (UNICEF), NIH, Drugs for Neglected Diseases initiative (DNDi), and the Paris Institut Pasteur. The Pasteur Institute of Iran is a member of the Pasteur International Network and has well-established links to Pasteur institutions in Paris, Tunisia, and Morocco. Increased investment in these types of research institutes will greatly advance research capacity building in the MENA region. Strengthening the connections between education, research, and the international scientific community can have a tremendous, positive effect on this region. By creating an active scientific atmosphere (for instance, via research training centers), established researchers will be motivated and young graduates will be attracted to careers in scientific research, improving the overall capability of the MENA region to control NTDs. In the coming years, geopolitical events taking place in the MENA region could promote the emergence or re-emergence of widespread NTDs. Now, more than ever, we need to re-examine the necessity to strengthen and expand existing programs for the control of NTDs in MENA, through improvement of mass treatment coverage for these diseases and identification of innovative ways to enhance R&D capacity in the region.

          Related collections

          Most cited references8

          • Record: found
          • Abstract: found
          • Article: not found

          First generation leishmaniasis vaccines: a review of field efficacy trials.

          First generation candidate vaccines against leishmaniasis, prepared using inactivated whole parasites as their main ingredient, were considered as promising because of their relative ease of production and low cost. These vaccines have been the subject of many investigations over several decades and are the only leishmaniasis vaccine candidates which have undergone phase 3 clinical trial evaluation. Although the studies demonstrated the safety of the vaccines and several studies showed reasonable immunogenicity and some indication of protection, an efficacious prophylactic vaccine is yet to be identified. Despite this overall failure, these trials contributed significantly to increasing knowledge on human leishmaniasis immunology. To provide a collective view, this review discusses the methods and findings of field efficacy trials of first generation leishmaniasis vaccine clinical trials conducted in the Old and New Worlds.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Leishmaniasis in the Middle East: Incidence and Epidemiology

            Leishmaniasis is a major health problem worldwide, with several countries reporting cases of leishmaniasis resulting in loss of human life or a lifelong stigma because of bodily scars. The Middle East is endemic for cutaneous leishmaniasis, with countries like Syria reporting very high incidence of the disease. Despite several countries establishing national control programs for containing the sandfly vector and treatment of infection, the disease continues to spread. In addition to the endemicity of the region for leishmaniasis, the Middle East has seen a great deal of human migration either for earning of livelihood or due to political upheaval in the region. These factors contribute to the spread and proliferation of the causative species Leishmania and its sandfly host. This review discusses the current epidemiological scenario in Iraq, Syria, Saudi Arabia, and Jordan, emphasizing the number of cases reported, vector species, Leishmania species, and treatment available. The data is primarily from WHO reports for each country and current and old literature.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Enhanced Protective Efficacy of Nonpathogenic Recombinant Leishmania tarentolae Expressing Cysteine Proteinases Combined with a Sand Fly Salivary Antigen

              Introduction Leishmaniasis is one of the greatest health challenges in nearly 98 countries, contributing to 2 million new clinical cases per year in tropical and subtropical regions of the globe [1]. The disease is transmitted by sandflies and is manifested in several clinical forms, mainly cutaneous leishmaniasis (CL), mucocutaneous leishmaniasis (MCL), and visceral leishmaniasis (VL) [2]. The geographical spread of the various clinical forms depends on vector availability. For instance, over 90% of CL cases occur in Afghanistan, Algeria, Brazil, Iran, Peru, Saudi Arabia and Syria; while, 95% of VL cases are found in Bangladesh, India, Nepal, Sudan, Ethiopia and Brazil [3]. High treatment costs, toxicity of drugs, and the constant emergence of parasite resistance highlight the need for a vaccine. Despite the observation that individuals with a healed primary Leishmania infection are protected against reinfection, no effective vaccine has been developed thus far. Lack of success may be due to our incomplete understanding of the control and regulation of immune responses during infection/reinfection and the mechanisms involved in the development of immune memory. In humans, acquired resistance to L. major infection is mediated primarily by cellular immunity, in particular antigen-specific type 1 T helper (Th1) cells. Similarly, Th1 dependent protection is observed in mouse experimental models of L. major infection. Most efforts for antigen identification have been focused on parasite proteins. More recently, it was shown that immunization with defined sand fly salivary proteins confers protection against leishmaniasis [4]. This suggests that salivary molecules can contribute to protection as a component of an anti-Leishmania vaccine. Live attenuated vaccines are the gold standard for protection against intracellular pathogens. Importantly, there have been some recent attempts using this approach for the development of Leishmania vaccines [5], [6]. Other approaches manipulate the Leishmania genome to engineer genetically modified parasites by introducing or eliminating particular virulence genes [7], [8], [9]. These approaches are powerful alternatives for the development of new generation vaccines against leishmaniasis. Nonpathogenic to humans Leishmania strains are also being assessed as promising vaccine tools [10]. Vaccination with a L. tarentolae recombinant strain expressing select immunogenic components of L. infantum, including the A2 and the cysteine proteinases A and B (CPA/CPB) genes as a tri-fusion conferred protection against L. infantum infection [11]. In the present study, we evaluated the efficacy of a new prime-boost vaccine combination consisting of a live recombinant nonpathogenic parasite and a vector salivary protein in eliciting a more powerful protective immunity against L. major infection. For this, we combined a recombinant L. tarentolae expressing the CPA/CPB cysteine proteinases with the immunogenic sand fly salivary molecule PpSP15 delivered as a DNA vaccine. We used different prime-boost regimens and evaluated the immunogenicity and protective effectiveness of this novel vaccine combination against L. major infection in both BALB/c and C57BL/6 mice. Materials and Methods Ethics statement All mouse experiments including maintenance, animals' handling program and blood sample collection were approved by Institutional Animal Care and Research Advisory Committee of Pasteur Institute of Iran (Research deputy dated October 2010), based on the Specific National Ethical Guidelines for Biochemical Research issued by the Research and Technology Deputy of Ministry of Health and Medicinal Education (MOHM) of Iran that was issued in 2005. Reagents All solutions were prepared using MilliQ ultrapure (Milli-QSystem, Millipore, Molsheim, France) and non-pyrogenic water to avoid surface-active impurities. G418, and Sodium dodecyl sulfate (SDS) were purchased from Sigma-Aldrich (Sigma, Deisenhofen, Germany). The material for PCR, enzymatic digestion and agarose gel electrophoresis were acquired from Roche Applied Sciences (Mannheim, Germany). Cell culture reagents including M199 medium, HEPES, L-glutamine, adenosine, hemin, gentamicin, DMEM and Schneider were purchased from Sigma (Darmstadt, Germany) and Gibco (Gibco, Life Technologies GmbH, Karlsruhe, Germany), respectively. Fetal Calf Sera (FCS) was purchased from Gibco (Gibco, Life Technologies GmbH, Karlsruhe, Germany). All cytokine kits were purchased from DuoSet R & D kits, (Minneapolis, USA). DNA constructs A 2.3 kb fragment content CPA/CPB/EGFP fused genes (with stop codons at the end of the EGFP ORF) was digested from pCB6-CPA/CPB/EGFP using SacI and BamHI and then cloned into the corresponding sites of pEGFP-N1 vector (Clontech, Palo Alto, CA) to provide the vector referred to as pEGFP-CPA/CPB/EGFP. After confirmation of the tri-fused gene through PCR and sequence determination, the pLEXSY-NEO2 vector (EGE-233, Jena bioscience, Germany) was used as an integrative vector to incorporate the CPA/CPB/EGFP fusion gene into the genome of the parasite. The CPA/CPB/EGFP was digested from pEGFP-CPA/CPB/EGFP using XhoI and XbaI and cloned into NheI and XhoI sites of the pLEXSY vector (XbaI and NheI are isoschizomers and make compatible sticky ends). For integration, the SwaI was used to linearize the vector at the 5′ and 3′ ends. Then the L. tarentolae (Tar II ATCC 30267) was grown in M199 5% inactivated fetal bovine serum (iFBS) to an optimal concentration. Parasite density was measured by counting the cells dissolved in Hyman's solution (HgCl2 0.5 g, NaCl 1 g, Na2SO4, 10H2O 11.5 g) using a hemocytometer. The pellet was resuspended in ice-cold electroporation buffer (21 mM HEPES, 137 Mm NaCl, 5 mM KCl, 0.7 mM Na2HPO4, 6 mM glucose; pH 7.5) to a final density of 108 parasites/ml, as recommended [12]. A total of 4.0×107 parasites/300 µl were mixed with 5–10 µg linearized DNA for stable transfection in a 0.2 cm electroporation cuvette (BioRad, USA) and stored on ice for 10 min. Electroporations were performed twice at 450 V, 500 µF using Bio Rad Gene Pulser Ecell device (Bio-Rad, USA) and the cell suspension was immediately put on ice for 10 min. Electroporated parasites were then transferred to 3 ml complete M199 media supplemented with 10% iFBS free of antibiotic and incubated at 26°C for 24 hours. Then, the live parasites were collected by centrifugation at 3000 rpm for 10 min at 4°C. Cells were subsequently transferred onto semi-solid plates of M199 medium containing 50% Noble agar (Difco, USA) and 50 µg/ml G418 (Gibco, Germany) and incubated at 26°C. The genotype of transfected parasites was confirmed by Southern blotting using the EGFPORF through incorporation of radiolabeled dCTP in a PCR reaction. In addition, genomic DNA obtained from transfected and wild type (WT) cells was amplified by PCR with specific primers to the upstream and downstream of the flanking region of 5′SSU. Forward primer (F3001) anneals upstream of the 5′SSU on WT genome and reverse primer (A1715) anneals to the backbone of the vector, downstream of 5′SSU and upstream of CPA/CPB/EGFP gene. The sequences for primer F3001 are: 5′ GAT CTG GTT GAT TCT GCC AGT AG 3′ and for primer A1715: 5′ TAT TCG TTG TCA GAT GGC GCA C 3′. The expression of CPA/CPB in the recombinant parasites was confirmed by RT-PCR, Western blot as well as flow cytometry. DNA vaccine constructs The gene coding for PpSP15 (NCBI accession number: AF335487) from the NH2 terminus to the stop codon was amplified from P. papatasi SP15-specific cDNA by PCR as reported previously [13] and cloned into the TOPO TA cloning vector PCRII (Invitrogen). The plasmid VR1020-SP15 was purified using the Endo Free Plasmid Mega kit according to the manufacturer's instructions (QIAGEN, Germany). Antigens and salivary gland preparation Frozen and thawed (F/T) L. major and L. tarentolae CPA/CPB/EGFP antigens were prepared from stationary phase promastigotes. Parasites were washed with PBS (8 mM Na2HPO4, 1.75 mM KH2PO4, 0.25 mM KCl, 137 mM NaCl) prior to 10 times exposition to liquid nitrogen and 37°C water bath alternately. The rCPA and rCPB were also prepared as previously reported [14]. Protein concentrations were measured with a BCA kit (PIERCE, Chemical Co., Rochford III). For preparation of salivary gland homogenate (SGH), P. papatasi females, Israeli strain, were used for dissection of salivary glands 3–7 d after emergence as previously described [15]. Briefly, salivary glands were disrupted by ultra-sonication and centrifuged at 10,000 g for 3 min and the resultant supernatant was dried in a Speed Vac (Thermo Scientific) and reconstituted before use in the listed experiments. Vaccination regimens and infectious challenge Female BALB/c (H-2d) and C57BL6 (H-2b) mice (6–8 weeks old, weighting 20±5 g) were purchased from the Pasteur Institute of Iran animal breeding facilities. All animals were housed in plastic cages with free access to tap water and standard rodent pellets in an air-conditioned room under a constant 12∶12 h light-dark cycle at room temperature and 50–60% relative humidity. Six groups of BALB/c or C57BL/6 mice (n = 20 per group) were vaccinated in different prime/boost modalities given three weeks apart in the right hind footpad (Table 1). These included, G1: vaccination with L. tarentolae CPA/CPB/EGFP+ and boosting with L. tarentolae CPA/CPB/EGFP+; G2: vaccination with VR1020-SP15 and boosting with L. tarentolae CPA/CPB/EGFP+ followed by VR1020-SP15 the day after; G3: vaccination with L. tarentolae CPA/CPB/EGFP+ followed by VR1020-SP15 the next day and boosting with L. tarentolae CPA/CPB/EGFP+ followed by VR1020-SP15 the next day; G4: Control group, vaccination with PBS; G5: vaccination and boosting with VR1020-SP15; G6: vaccination and boosting with L. tarentolae EGFP+. L. major EGFP+ (MRHO/IR/75/ER) parasites were used for the infectious challenge and were kept in a virulent state by continuous passage in BALB/c mice. The promastigotes were cultured in M199 medium supplemented with 5% iFBS and 40 mM HEPES, 0.1 mM adenosine, 0.5 µg/ml hemin, 2 mM L-glutamine and 50 µg/ml gentamicin at 26°C. For mice challenge, a total of 2×105 stationary phase promastigotes were injected subcutaneously in the left hind footpad 3 weeks after the booster immunization. For G2, G3, G4 and G5, 0.5 pair of sand fly SGH was mixed with parasites and used for challenge. 10.1371/journal.pntd.0002751.t001 Table 1 Vaccination regimens in BALB/c and C57BL/6 mice models. Groups Prime Boost Challenge Modality Group 1 N = 20 L. tarentolae (CPA/CPB/EGFP) 2×107 L. tarentolae (CPA/CPB/EGFP) 2×107 L. major Footpad s.c. 2×105 rLive/rLive vaccine Group 2 N = 20 VR1020-SP15 20 µg/mice L. tarentolae (CPA/CPB/EGFP) 2×107 VR-SP15 (next day) L. major+SGH Footpad s.c. 2×105 DNA/rLive+DNA vaccine Group 3 N = 20 L. tarentolae (CPA/CPB/EGFP) 2×107 VR1020-SP15 (next day) L. tarentolae (CPA/CPB/EGFP) 2×107 VR1020-SP15 (next day) L. major+SGH Footpad s.c. 2×105 rLive+DNA/rLive+DNA vaccine Group 4 N = 20 PBS PBS L. major+SGH Footpad s.c. 2×105 CONTROL Group 5 N = 20 VR1020-SP15 20 µg/mice VR1020-SP15 20 µg/mice L. major+SGH Footpad s.c. 2×105 DNA/DNA vaccine Group 6 N = 20 L. tarentolae 2×107 L. tarentolae 2×107 L. major Footpad s.c. 2×105 Live/Live vaccine CONTROL Cytokine assays before and after challenge with L. major The profile of cytokine production in the groups vaccinated with L. tarentolae CPA/CPB/EGFP+ (G1) and a combination of L. tarentolae CPA/CPB/EGFP+ and VR1020-SP15 (G2, and G3) and the PBS-immunized control group G4 in both BALB/c and C57BL/6 mice was measured before challenge and at 3 and 10 weeks post challenge in two independent repeats. Briefly, at each time point, 4 mice from each group were sacrificed. Their spleen was treated with a tissue grinder and red blood cells were lysed for 5 minutes using the ACK lyses buffer (NH4Cl 0.15M, KHCO3 1 mM, Na2EDTA 0.1 mM). Splenic cells were then washed, put in culture at 3.5×106 cells/ml and exposed to recombinant antigens rCPA (10 µg/ml) and rCPB (10 µg/ml), F/T lysate of L. major (15 µg/ml), L. tarentolae harboring cysteine proteinase genes of interest (25 µg/ml), and SGH (2pairs/ml). Cell culture supernatants were collected after 24 hours for IL-2 and TNF-α assays and 72 hours later for IFN-γ and IL-4 assays. Cytokine measurements were performed by Sandwich ELISA using the DuoSet R & D kits as per the manufacturer's instructions. The minimum detection limit is 2 pg/ml for mouse IFN-γ and IL-4, 3 pg/ml for IL-2 and 5 pg/ml for TNF-α. All measurements were run in duplicates for two independent experiments. Concanavalin A (Con A; 5 µg/ml) was used in all experiments as a positive control. Determination of Ag-specific antibody responses For the groups vaccinated with L. tarentolae CPA/CPB/EGFP+ (G1) and a combination of L. tarentolae CPA/CPB/EGFP+ and VR1020-SP15 (G2, and G3) and the PBS-immunized control group G4, mice were bled to obtain serum for determination of antibody responses. The serum sample obtained from each mouse was analyzed by ELISA for specific IgG1 and IgG2a isotype responses three weeks after booster immunization (against F/T lysate of L. tarentolae CPA/CPB/EGFP+ (10 µg/ml) and SGH (2pair/ml) and at 5 weeks after challenge against F/T lysate of L. major (10 µg/ml) and SGH (2pair/ml). Briefly, 96-well plates (Greiner) were coated with each antigen overnight at 4°C. Plates were blocked with 100 µl of 1% BSA in PBS at 37°C for 2 h to prevent nonspecific binding. Sera (1∶100) were added and incubated for 2 h at 37°C. After three washes, goat anti-mouse IgG1-HPR (1∶10,000, Southern Biotech, Canada) or goat anti-mouse IgG2a-HPR (1∶10,000, Southern Biotech, Canada) were added and incubated for 2 h at 37°C. After four washes, plates were incubated for 30 min at 37°C with Peroxidase Substrate System (KPL, ABTS) as substrate. Reactions were stopped with 1% SDS and the absorbance was measured at 405 nm. Parasite burden The parasite load in different groups of BALB/c and C57BL/6 mice (G1, G2, G3, G4, G5 and G6) were determined by the limiting dilution assay at 3 and 10 weeks post challenge [16]. Briefly, at each time point 4 mice from each group were taken randomly, sacrificed and the lymph nodes (LN) were excised and weighed. After homogenizing, 20 different serial dilutions (10−1 to 10−20) were prepared in Schneider's Drosophila medium supplemented with 10% iFBS and gentamicin (0.01%). Diluted cells were cultured in 96 well plates in duplicate and investigated 7 and 14 days later for positive wells. The parasite load was calculated using the following formula: −Log10 (last dilution with live parasites/weight of homogenized LN). Imaging of infection intensity of EGFP-transfected L. major in the footpad of BALB/c mice To demonstrate the in vivo level of infection, the infected footpad (FP) was imaged 10 weeks after challenge with the KODAK Image Station 4000 Digital Imaging System. Briefly, six BALB/c mice from each group (G1, G2, G3, G4, G5 and G6) were treated with a depilatory substance (Nair) to remove hair from their FPs to reduce background auto fluorescence. Afterward mice were temporarily anesthetized intraperitoneally with a mixture of xylazine 2% (7.5 µl), Ketamine 10% (30 µl) and saline solution (260 µl) per mouse and then imaged. Pixel counting and measurement of the lesions were performed using the KODAK molecular image software version 5.3. Measurements were reported as “Net intensity”, a quantitative measurement defined as the number of green pixels in a given area multiplied by the average intensity of each pixel minus the background intensity. Statistical analysis Statistical analysis was performed using Graph-Pad Prism 5.0 for Windows (San Diego, California). Depending on data passing normality tests, ANOVA or Mann-Whitney U tests were computed. P values less than 0.05 were considered significant. The specific test employed is indicated in each figure. Results Generation of a recombinant L. tarentolae expressing the CPA/CPB/EGFP tri-fusion gene Expression of the 2.3 kb CPA/CPB/EGFP tri-fusion genes in L. tarentolae is under the regulatory control of the rRNA Pol I promoter. We integrated the CPA/CPB/EGFP fragment flanked by 5′(∼860 bp) and 3′SSU (∼1080 bp) sequences into the rRNA locus of L. tarentolae (Figure 1A). The recombinant L. tarentolae strain expressing CPA/CPB/EGFP genes displayed a normal morphology (a drop-like shape) with a normal length of the flagellum comparable to that of the wild type strain. EGFP expression and intensity were verified by visualization using an epifluorescence microscope. The EGFP was attached to the C-terminal end of CPB and fluorescence is distributed through the whole cytoplasm (Figure 1B). Confirmation of CPA/CPB/EGFP expression at the level of RNA and protein was verified using RT-PCR and western blot, respectively (data not shown). 10.1371/journal.pntd.0002751.g001 Figure 1 Schematic structure of the 5′SSU-CPA/CPB/EGFP-3′SSU cassette linearized with SwaI to integrate it into the SSU region of the L. major genome (A). Microscopic analysis of L. tarentolae stably expressing CPA/CPB/EGFP into the ribosomal locus (B). Pre-challenge evaluation of the immunogenicity of live recombinant L. tarentolae CPA/CPB/EGFP combined with PpSP15 in three different regimens in BALB/c and C56BL/6 mice A major requirement of vaccines is to protect the majority of a population that normally displays a high diversity in MHC haplotypes. It is known that L. major causes a non-healing cutaneous infection in susceptible BALB/c mice characterized by progressive skin lesions and visceralization of the parasites to the spleen [17], [18]. In contrast, C57BL/6 mice are naturally resistant against L. major and the infection normally causes transient symptoms and is self-healing [18]. Therefore, we evaluated the immune response in both BALB/c (H-2d) and C57BL/6 (H-2b) mice in the groups vaccinated with L. tarentolae CPA/CPB/EGFP+ (G1), combination of L. tarentolae CPA/CPB/EGFP+ and VR1020-SP15 (G2, and G3) and the PBS-immunized control group G4 (Table 1). It has been shown that IFN-γ and TNF-α are important parameters for vaccine evaluation since they synergize their capacity to mediate killing of pathogens. Furthermore, IL-2 also enhances the expansion of T cells, leading to a more efficient effector responses [19]. Since these effector cytokines mediate protection, we evaluated antigen specific immune responses three weeks after booster immunization by measuring the production of IFN-γ, IL-4, IL-2 and TNF-α in the supernatant of splenocytes in response to rCPA/rCPB or F/T lysate of L. tarentolae CPA/CPB/EGFP. In susceptible BALB/C mice, the levels of IFN-γ production by splenocytes after rCPA/CPB stimulation were significantly higher (p 0.05). For resistant C57BL/6 mice, splenocytes produced significantly higher levels of IFN-γ and IL-4 in the three vaccinated groups (G1, G2, G3) as compared to control group G4 when stimulated with either rCPA/CPB or L. tarentolae CPA/CPB/EGFP (Figure 3A, p<0.05). Nevertheless, the level of IL-4 was lower than that of IFN-γ in all vaccinated groups (Figure 3A). Groups G1, G2 and G3 produced significantly higher levels of IL-2 compared to G4 (p<0.05) when stimulated with rCPA/CPB or L. tarentolae CPA/CPB/EGFP (Figure 3A). As for TNF-α, it was only produced upon stimulation with rCPA/CPB where G1, G2 and G3 showed significantly higher levels in comparison to G4 (Figure 3A). 10.1371/journal.pntd.0002751.g003 Figure 3 Cellular and humoral immune responses by splenocytes of C57BL/6 mice measured after last immunization. A) Cytokine production from the splenocytes of 4 mice from the different groups at 3 weeks after last immunization. Cells cultured in duplicate in the presence of recombinant CPA/CPB or freeze-thawed recombinant L. tarentolae. Culture supernatants were assayed for IFN-γ, IL-4, IL-2 and TNF-α production by ELISA. Each bar represents the Mean±SD of 4 mice per group. B) Antigen specific antibody responses against L. tarentolae CPA/CPB/EGFP and SGH before challenge. Data were generated as Mean±SD for sera of individual mice within their respective vaccination group (n = 8–10). The asterisk indicates the significant difference between values at the indicated time points as determined by the Mann Whitney U test (p<0.05 denoted as *), compared to the control group (G4). Results are representative of two independent experiments. The three vaccinated groups produced significantly higher levels of IgG2a against L. tarentolae CPA/CPB/EGFP compared to control G4 (Figure 3B). The level of IgG1 was only significantly higher in G3 as compared to control G4 (p<0.05). Furthermore, there were no significant differences between IgG1 and IgG2a levels against SGH in the three tested groups G2, G3 and G4 (Figure 3B). Evaluation of vaccine efficacy in susceptible BALB/c mice All six groups of vaccinated and control BALB/c mice (Table 1) were challenged with 2×105 late-stationary phase L. major GFP+ promastigotes in their left footpads in the presence (G2, G3, G4, G5) or absence (G1, G6) of SGH. Weekly measurements showed a sharp increase in footpad swelling in the control groups G4 and G6 at weeks 8, 9 and 10 that was significantly larger than that observed in groups G1, G2, G3 and G5 (Figure 4A p<0.05). As a main parameter, the parasite burden was measured in the lymph nodes of all six groups at 3 and 10 weeks post challenge using a limiting dilution assay (Figures 4B). Three weeks after challenge (3WAC), groups G1, G2, G3 and G5 showed a significantly lower parasite load than groups G4 and G6 (Figure 4B) with G2 and G5 showing the lowest lymph node parasite burden (Figure 4B). At the end of week 10, the parasite burden of groups G1, G2, G3 and G5 remained significantly lower (p<0.05) compared to groups G4 and G6 (Figure 4B). In addition, both G2 and G3 has significantly lower parasite load in respect to G1 and G5 (p<0.05). In vivo imaging of fluorescent parasites in the footpad 10 weeks after challenge (10WAC) shows a significant reduction in the level of fluorescence intensity in the footpad of the vaccinated groups G1, G2 and G3 as compared to the control group G4 (Figures 4C–D). Group G3 had the lowest fluorescence intensity with two mice showing no GFP fluorescence (Figure 4C–D). Moreover, the fluorescence intensity of group G3 was significantly lower in comparison to groups G1, G4, G5 and G6 (p<0.05) but was not statistically significant from that of group G2 (Figure 4D). 10.1371/journal.pntd.0002751.g004 Figure 4 The course of infection with L. major GFP+ in BALB/c mice vaccinated with different modalities. BALB/c mice were immunized in the right footpad with L. tarentolae CPA/CPB/EGFP (G1, rLive/rLive); primed with DNA PpSP15 and boosted with L. tarentolae CPA/CPB/EGFP and DNA PpSP15 (G2, DNA/rLive+DNA); primed and boosted with both L. tarentolae CPA/CPB/EGFP and DNA PpSP15 (G3, DNA+rLive/DNA+rLive); injected with PBS as control (G4), vaccination and boosting with VR1020-SP15 (G5, DNA/DNA); G6: vaccination and boosting with L. tarentolae EGFP+ (G6, control Live/Live). All animals were challenged with stationary phase L. major (2×106/mice) plus SGH (0.5 pair) in the left footpad except for G1 and G6, which received only L. major. A) The footpad swelling represents the Mean±SD of 12 mice per group; Asterisks indicate statistical significance (Mann Whitney U test, *p<0.05) compared to the control group (G4). B) Parasite burden per lymph node in all groups at 3 and 10 weeks post challenge (WAC). Each data point represents the Mean ±SD of 4 lymph nodes per group; statistics were carried out by ANOVA. C) Photographs of mouse footpads infected with fluorescent L. major for G1–G6. D) Net intensity by fluorescence imaging at 10WAC; statistical differences was determined by the Mann Whitney U test (p<0.05 denoted as *). Values of two independent experiments are shown in the figure. For assessment of the immune response after challenge, we focused on the groups vaccinated with L. tarentolae CPA/CPB/EGFP+ (G1) and a combination of L. tarentolae CPA/CPB/EGFP+ and VR1020-SP15 (G2, and G3) compared to the PBS-immunized control group G4. Splenocytes stimulated with L. major F/T antigen at 3WAC show that groups G1, G2 and G3 produced significantly higher levels of IFN-γ compared to control group G4 (Figure 5A, p<0.05). Though group G3 produced higher levels of IFN-γ compared to group G2, it also produced significantly higher levels of IL-4 (p<0.05) as compared to groups G1, G2 and G4 (Figure 5B). The difference in the levels of these two cytokines became less pronounced at 10WAC (Figure 5B). All vaccinated groups showed a positive IFN-γ/IL-4 ratio and group G2 had the highest IFN-γ/IL-4 ratio at 3WAC indicative of a Th1 response (Figure 5C). At 10WAC, group G3 had the highest ratio of IFN-γ/IL-4 (p<0.05) in comparison to G1, G2, G4. With regard to IL-2 production, only G2 produced significantly higher levels of this cytokine as compared to G1, G3 and G4 at 3WAC and 10WAC (Figure 5D). TNF-α production was similar at in all vaccinated and control groups at 3WAC but it was significantly higher in the three vaccinated groups (G1, G2 and G3) compared to control group G4 at 10WAC (Figure 5E, p<0.05). 10.1371/journal.pntd.0002751.g005 Figure 5 The immune response post-challenge in BALB/c mice vaccinated with different modalities. A–E, Cytokine production by splenocytes at 3 and 10 weeks after challenge (WAC). Single cell suspensions were prepared from splenocytes of 4 mice after infectious challenge. Cells were cultured in duplicate in the presence of freeze-thawed L. major +SGH for G2, G3 and G4 and with only L. major for G1. Culture supernatants were assayed for the level of IFN-γ (A), IL-4 (B), ratio of IFN-γ to IL-4 (C), IL-2 (D) and TNF-α (E) production by ELISA. Each bar represents the Mean±SD of 4 mice per group. The asterisk indicates the significant difference between values at the indicated time points as determined by Mann Whitney U test compared to the control group (G4). Antigen-specific antibody responses against L. major (F) and SGH (G) five weeks after challenge. Data represent the serum IgG1 and IgG2a levels of individual mice within their respective vaccination group (n = 8–10). Statistical analysis was carried out by Mann Whitney U test comparing vaccination groups against G4 as control (p<0.05 denoted as *). Results are representative of two independent experiments. The specific antibody response against L. major in BALB/c mice was measured in the above-mentioned groups at 5 weeks after challenge. Groups G2 and G3 displayed the highest level of IgG2a and IgG1 antibodies to Leishmania compared to group G1, and control group G4 (p<0.05, Figure 5F). The low levels of IgG1 and IgG2a antibodies to Leishmania were similar in groups G1 and G4 (Figure 5F). Regarding anti-sand fly saliva antibodies, the levels of IgG2a antibodies were significantly higher in groups G2 and G3 compared to control group G4 (Figure 5G). Furthermore, the ratio of saliva-specific IgG2a/IgG1 was greater in groups G2 and G3 (Figure 5G). Evaluation of vaccine efficacy in resistant C57BL/6 mice In C57BL/6 mice the increase in footpad swelling was similar between groups G1, G4 and G6 (Figure 6A). There was a significant decrease (p<0.05) in footpad swelling in groups G2 and G3 in comparison to groups G1, G4, G5 and G6 (Figure 6A). Measurements of parasite burden from lymph nodes at 3 and 10 weeks post-challenge showed that, group G2 had significantly the lowest parasite burden (p<0.05) as compared to groups G1, G3, G4, G5 and G6 (Figure 6B). We also observed at 10WAC a significant decrease in parasite burden in G1, G3 and G5 as compared to control group G4 as well as G6. In addition, the level of parasite burden in G6 is also significantly lower than G4 (p<0.05) (Figure 6B). Overall, these data demonstrate that in C57BL/6 mice priming with VR1020-SP15 and boosting with a combination of the live vaccine and VR1020-PpSP15 elicited a higher protective efficacy than the two other regimens in controlling footpad swelling and parasite propagation up to 10 weeks post-challenge (Figure 6A, B). Although we were able to detect the swelling in the footpad of all groups, we were unable however, to determine the fluorescent intensity by imaging in the footpad of C57BL/6 mice as it was done for BALB/c. In fact, in C57BL/6 mice resistant strain, the level of parasite propagation in the footpads of all groups was limited (below the instrument detection limit). 10.1371/journal.pntd.0002751.g006 Figure 6 The course of infection with L. major GFP+ in C57BL/6 mice vaccinated with different modalities. C57BL/6 mice were immunized in the right footpad with L. tarentolae CPA/CPB/EGFP (G1; rLive/rLive); primed with DNA PpSP15 and boosted with L. tarentolae CPA/CPB/EGFP and DNA PpSP15 (G2; DNA/rLive+DNA); primed and boosted with L. tarentolae CPA/CPB/EGFP and DNA PpSP15 (G3; DNA+rLive/DNA+rLive); injected with PBS as control (G4), vaccination and boosting with VR1020-SP15 (G5, DNA/DNA); G6: vaccination and boosting with L. tarentolae EGFP+ (G6, control Live/Live). All animals were challenged with stationary phase L. major (2×106/mice) plus SGH (0.5 pair) in the left footpad except for G1 and G6 which only received L. major. A) The footpad swelling represents the Mean±SD of 12 mice per group. Asterisks indicate statistical significance (Mann Whitney U test, p<0.05) compared to the control group (G4). B) Parasite burden per lymph node in all groups at 3 and 10 weeks post challenge (WAC). Each data point represents the Mean ±SD of 4 lymph nodes per group. Statistics were carried out by ANOVA (p<0.05 denoted as *). Values of two independent experiments are shown in the figure. Similar to BALB/c mice, we focused on the groups vaccinated with L. tarentolae CPA/CPB/EGFP+ (G1) and a combination of L. tarentolae CPA/CPB/EGFP+ and VR1020-SP15 (G2, and G3) compared to the PBS-immunized control group G4 for assessment of the immune response after challenge. Splenocytes stimulated with L. major F/T antigen at 3WAC show that groups G2 and G3 had higher levels of IFN-γ compared to control group G4 (Figure 7A). At 10WAC, IFN-γ production was similar among the vaccinated groups G1, G2 and G3 and was significantly higher than control group G4 (Figure 7A). The levels of IL-4 were lower in group G2 as compared to group G3 at 3WAC (Figure 7B, p<0.05), however this cytokine decreased significantly 10WAC in group G3 compared to groups G1, G2 and G4. Similar to BALB/c mice, the ratio of IFN-γ/IL-4 was higher in groups G1, G2 and G3 compared to the control group G4, particularly at 10WAC (Figure 7C). It is worth to mention that G3 has significantly the highest ratio in compare to G1 and G2 (p<0.05). As for IL-2, its production was significantly higher (p<0.05) in groups G2 and G3 compared to groups G1 and G4 at 3WAC, but no statistical significance was observed among the groups at 10WAC (Figure 7D). Importantly, the induction of TNF-α was significantly higher in Group G2 as compared to control group G4 at 10WAC and was 2-folds higher compared to G1 and G3 (Figure 7E). 10.1371/journal.pntd.0002751.g007 Figure 7 The immune response post-challenge in C57BL/6 mice vaccinated with different modalities. A–E, Cytokine production by splenocytes at 3 and 10 weeks after challenge. Single cell suspensions were prepared from splenocytes of four C57BL/6 mice at 3 and 10 weeks after infectious challenge. Cells were cultured in duplicate in the presence of freeze-thawed L.major+SGH for G2, G3 and G4 and with only L. major for G1. Culture supernatants were assayed for the level of IFN-γ (A), IL-4 (B), ratio of IFN-γ to IL-4 (C), IL-2 (D) and TNF-α (E) production by ELISA. Each bar represents the Mean±SD of 4 mice per group. Data were analyzed by the Mann Whitney test. Asterisks indicate the statistical differences between values at the indicated time points as compared to the control group (G4). Antigen-specific antibody responses against L. major (F) and SGH (G) five weeks after challenge. Data represent the serum IgG1 and IgG2a levels of each individual mouse within their respective vaccination group (n = 8–10). Statistical analysis was carried out by Mann Whitney U test (p<0.05 denoted as *). Results are representative of two independent experiments. Antibodies against L. major in vaccinated groups G1, G2 and G3 showed significantly higher levels of IgG2a in comparison to group G4 (p<0.05, Figure 7F). Furthermore, the levels of IgG1 were significantly lower than IgG2a in these three vaccinated groups (G1, G2 and G3) in comparison to G4 (p<0.05, Figure 7F). Overall, the antibody response to SGH was low (Figure 7G). With the exception of group G2 that produced significantly higher levels of IgG2a compared to groups G3 and G4, the antibody response to SGH was mixed (Figure 7G). Discussion Despite substantial progress in fundamental Leishmania research, there are many unanswered questions concerning pathogenesis of the disease and the acquisition of protective immunity against reinfection. In this respect, immunization with live attenuated strains as a vaccine tool to induce a protective immune response in the host has a long tradition [20]. The major drawback of this approach is that under certain circumstances, the strains may gain virulence and become pathogenic again. To overcome this problem, subunit vaccines, instead of the whole organism, emerged as a vaccination strategy [21]. A number of parasite antigens have been tested for their potential to induce anti-Leishmania responses. The most extensively studied antigens using a wide range of adjuvants and delivery systems are GP63, LACK, CPs, and the poly-antigen Leish111f [22], [23], [24]. In an attempt to engage Leishmania infection at an early stage, salivary proteins of the sand fly have also been evaluated for vaccination. Studies in mice, hamsters and dogs showed promising results with the induction of Th1-like responses and long-term protection against both cutaneous and visceral infections using these salivary proteins [4], [8]. Here, we describe the outcome of a new vaccination strategy with different modalities using a live recombinant nonpathogenic L. tarentolae vaccine expressing CPA/CPB/EGFP combined to a DNA vaccine containing the cDNA for PpSP15, the predominant 15 kDa salivary protein from the sand fly P. papatasi. Our target parasite antigens are cysteine proteinases, which are conserved among different Leishmania species and are highly immunogenic. L. tarentolae, the lizard protozoan parasite, has been previously introduced by Breton et al. [25] as a candidate vaccine against visceral leishmaniasis. Furthermore, we have demonstrated that a recombinant L. tarentolae strain expressing the L. donovani A2 gene elicited a strong protective immunity against virulent L. infantum challenge [26]. Recently, we have shown that vaccination with L. tarentolae expressing A2/CPA/CPB induced a strong parasite-specific Th1 response and conferred protection against L. infantum challenge in BALB/c mice [11]. As for PpSP15, it was shown previously to protect vaccinated C57BL/6 mice challenged with parasites plus SGH [15], [27]. A major requirement of vaccines in general, is that they are able to protect the majority of a population, which normally displays a high diversity in MHC haplotypes. For this reason, we tested the efficacy of the recombinant live L. tarentolae expressing CPA/CPB/EGFP candidate vaccine combined to PpSP15 DNA in eliciting protective immune responses in two different strains of mice. While BALB/c mice develop progressive lesions upon infection with L. major, C57BL/6 mice are naturally resistant and the infection normally causes transient symptoms (contained lesion development and visceralization) and is self-healing. In this study, L. major IR75 was used for an infectious challenge because it is more virulent in comparison to L. major 39 and the Friedlin strain (Modabber F, personal communication). Both strains of mice showed the strongest protective effect following immunization with a prime/boost regimen based on PpSP15 DNA and recombinant L. tarentolae (groups G2 and G3) demonstrating an enhanced vaccine efficacy compared to the sole use of L. tarentolae CPA/CPB/EGFP (G1) or PpSP15 DNA (G5). While group G3 showed a more potent immune response in susceptible BALB/c mice, group G2 showed the strongest immunogenicity in C57BL/6 mice and it was the best group in controlling parasite growth in the lymph nodes of both mice strains. In both strains of mice, immunization with PpSP15 as a DNA vaccine combined to L. tarentolae CPA/CPB/EGFP showed considerable level of protection as demonstrated by footpad thickness measurements and parasite burden. This demonstrated for the first time the effectiveness of co-immunization of a sand fly salivary protein, PpSP15, with live L. tarentolae CPA/CPB/EGFP in controlling the disease. In the case of BALB/c mice, the effect of Live L. tarentolae CPA/CPB/EGFP is less pronounced although we observed a significantly lower parasite burden in G2 and G3 compared to G1, G4, G5 and G6. Inclusion of PpSP15 DNA as a vaccine may be relevant at two levels: i) as an inducer of adaptive immunity, thus reducing lesion pathology and parasite propagation and ii) as an potential enhancer of innate immunity due to the intrinsic properties of this molecule that may contribute to the control of intracellular growth of L. major. Furthermore, there are extensive data showing that live L. major plus CpG DNA prevents lesion development and causes the specific induction of Th17 cells, which enhances the development of a protective cellular immunity against the parasite [28], [29]. Data presented by Mendez et al. [30] suggest that a vaccine combining live pathogens with immunomodulatory molecules may strikingly modify the natural immune response to infection in an alternative manner to that induced by killed or subunit vaccines. Therefore, it may be possible that PpSP15 working as an immunomodulatory molecule and enhancing the development of a protective cellular immunity against the parasite. Comparing the data obtained in C57BL/6 with BALB/c, the highest level of TNF-α production, indicative of a Th1 response, was seen with group G2 at 10WAC although there were no significant differences in IFN-γ production. Of note, we only checked four key cytokines to demonstrate the immunogenicity of each vaccine modality using the live recombinant L. tarentolae. We acknowledge the need to further investigate the role or contribution of other cytokines when studying live parasite vaccines. Our future efforts should be also focused on the analysis of the immunological memory and the factors that could correlate with the size of the memory pool using these vaccine strategies. One of these aspects is the induction of CD8 T+ cell responses, which remains to be elucidated. The concept of using live vaccination against leishmaniasis is not new. Actually, the inoculation of live parasites to produce a lesion that heals, named leishmanization, has been the only vaccination strategy implemented at a large scale because it provides lifelong protection against the development of additional lesions [31]. However, this approach was discontinued because of raised non-healing or slow healing lesions in several human cases [31], [32]. During the last few years, several attenuated strains of Leishmania have been developed. As an alternative, various defined genetically modified parasites have been achieved using a gene targeted disruption strategy through homologous recombination. One of the first examples was the in vivo evaluation showed that the dhfr-ts−/−parasites survived but were unable to establish a persistent infection or to cause disease even in the most susceptible mouse strains [33]. Other examples such as LPG2−/− parasites protected highly susceptible BALB/c mice against a L. major virulent challenge even in the absence of a strong Th1 response [34], [35]. In contrast to L. major mutants, L. mexicana LPG2−/− mutants retained their virulence for macrophages and mice [35], which suggested that different Leishmania species possess alternative virulence repertoires to interact with their host. Therefore, major safety constrains, such as a possible reversion to virulence or reactivation in immunosuppressed individuals, are still among the limiting factors against the use of such vaccines. In contrast to all of the above-mentioned approaches, L. tarentolae is non-pathogenic to humans and can be used in immunocompromised individuals. As such, recombinant L. tarentolae could offer more advantages for vaccine development not only against Leishmania, but also against other pathogens. A recombinant L. tarentolae expressing HIV-1 Gag protein induced strong cell-mediated immunity in mice and decreased HIV-1 replication in an ex vivo system, suggesting that this species can be applied as a promising live vaccine against intracellular pathogens [10]. Recently, a recombinant L. tarentolae strain expressing HPV-E7 antigen-green fluorescent protein (GFP) was developed and showed a potential as a live vaccine against HPV infection [36]. Additionally, modification of, and insertion into, the genome of L. tarentolae can be done easily and there is no insert size limitation making it a versatile tool for vaccine development. Our data clearly demonstrate that group G2 (prime with PpSP15 DNA and boost with L. tarentolae CPA/CPB/EGFP+PpSp15 DNA) has the lowest level of parasite propagation at 3WAC in both mice strains and at 10WAC in C57BL/6 mice. Therefore, apart from the specific immunogenicity of PpSP15, this salivary protein may have an immunomodulatory role that in combination with a live vaccine potentially enhances its efficacy against Leishmania. In summary, the present study suggests that this new approach that combines a prime-boosting strategy using recombinant L. tarentolae with a sand fly salivary protein offers a promising platform for developing a more effective vaccine against leishmaniasis.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Negl Trop Dis
                PLoS Negl Trop Dis
                plos
                plosntds
                PLoS Neglected Tropical Diseases
                Public Library of Science (San Francisco, CA USA )
                1935-2727
                1935-2735
                27 August 2015
                August 2015
                : 9
                : 8
                : e0003695
                Affiliations
                [1 ]Department of Immunotherapy and Leishmania, Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
                [2 ]Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
                [3 ]Director Office, Pasteur Institute of Iran, Tehran, Iran
                University of Notre Dame, UNITED STATES
                Author notes

                The authors have declared that no competing interests exist.

                Article
                PNTD-D-14-02110
                10.1371/journal.pntd.0003695
                4551800
                26313269
                c861802e-5590-4c81-9e45-74c4d2458e39

                This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication

                History
                Page count
                Figures: 0, Tables: 0, Pages: 4
                Funding
                The authors received no specific funding for this work.
                Categories
                Editorial

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article