Expert Review of Vaccines

ISSN: 1476-0584 (Print) 1744-8395 (Online) Journal homepage: http://www.tandfonline.com/loi/ierv20

Delivery systems for Leishmania vaccine development Sima Rafati, Elham Gholami & Farnaz Zahedifard To cite this article: Sima Rafati, Elham Gholami & Farnaz Zahedifard (2016): Delivery systems for Leishmania vaccine development, Expert Review of Vaccines, DOI: 10.1586/14760584.2016.1157478 To link to this article: http://dx.doi.org/10.1586/14760584.2016.1157478

Accepted author version posted online: 24 Feb 2016.

Submit your article to this journal

Article views: 3

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=ierv20 Download by: [137.189.171.235]

Date: 01 March 2016, At: 03:00

Publisher: Taylor & Francis Journal: Expert Review of Vaccines DOI: 10.1586/14760584.2016.1157478 Delivery systems for Leishmania vaccine development

Authors: Sima Rafati*, Elham Gholami and Farnaz Zahedifard

Downloaded by [] at 03:00 01 March 2016

Pasteur Institute of Iran - Department of Immunotherapy and Leishmania Vaccine Research, Tehran, Iran (the Islamic Republic of)

Corresponding author: Sima Rafati [email protected] Summary Leishmaniasis is a neglected disease and is endemic in tropical and sub-tropical areas worldwide. Lifelong immunity after recovery indicates that vaccination could be a promising approach to overcome the disease. Although different antigens have been successfully tested against all clinical forms, none of them have been shown to fulfill the safety and efficiency requirements for human applications. Hence, strong vehicles are needed to carry antigens of interest and potentiate its presence in the body. So far, various live or chemical carriers have been applied to reinforce the immunological effects of ideal antigens. In the current review, the recent attempts in this field have been summarized. Key words: Leishmaniasis, Vaccination, Live delivery system, Chemical delivery system, Nano/Microparticle, Protective immune responses

Downloaded by [] at 03:00 01 March 2016

Introduction Leishmaniasis is a parasitic disease caused by a protozoa parasite from over 20 Leishmania species. The disease is transmitted to humans by the bite of infected female sand flies. There are three main forms of the disease including visceral (VL), cutaneous (CL) and mucocutaneous (MC) leishmaniasis. This disease being one of the most neglected tropical diseases in the world, affects different populations in tropical and sub-tropical areas. According to WHO reports, every year the world faces 300,000 new cases of visceral form, leading to 20,000 deaths. Over 90% of new VL cases occur in 6 countries, namely Bangladesh, Brazil, Ethiopia, India, South Sudan and Sudan. in the case of cutaneous form, over 250,000 new cases are diagnosed in undeveloped and developing countries and 310 million individuals are at risk of infection. Over two thirds of new CL cases occur in 6 countries: Afghanistan, Algeria, Brazil, Colombia, Iran, and Syria. Beside the mortality of visceral form, victims of cutaneous and mucocutaneous forms face complications of mixed infection, disfiguration and disease recurrence in their life time (http://www.who.int/leishmaniasis/en/). One approach to prevent the disease is vaccination. In recent decades, many attempts have been made to test different vaccine candidates against various forms of leishmaniasis. Until now, only one approach has been applied to clinical trial phase at least for humans (LEISH-F3+GLA-SE) [1]. HIV coinfection in immuno-compromised patients increases the importance of finding effective vaccines against leishmaniasis. The vast majority of vaccination strategies have focused on finding a solution for this major health problem. Through time it has been understood that due to the complex nature of Leishmania infection, vaccination strategies should be potentiated with different delivery systems to inhibit the immediate degradation of antigen in the body or play a maintenance role in delivering the immunogen to the right site in a longer time. The aim of this review is to discuss and highlight different delivery systems including, live and chemical delivery systems which have been used to strengthen the immune response for vaccine development against leishmaniasis (Fig. 1, 2).

Live delivery systems: Application of live vaccination strategies is considered as one of the most promising approaches for a successful vaccination program. Researchers believe that immunological response against live microbes harboring special antigens would be strong enough to generate persistent immunity against diseases such as leishmaniasis. Natural active immunity against the disease is achieved by maintenance of some live parasites in the body (unsterile immunity) following self-limited infection. Although safety and practical issues of such vaccines are always the subject of controversies, researchers have

tried different strategies to deliver vaccine candidates through live organisms. In this section, viral, bacterial and parasitic vehicles as live delivery systems are reviewed.

Downloaded by [] at 03:00 01 March 2016

Viral delivery vehicles: Viruses could be engineered to express different exogenous antigens on their surface to harbor immunogenic molecules as inducers of the immune system. Three different vaccination studies have been performed against cutaneous leishmaniasis using Vaccinia as the vehicle. The most famous Leishmania antigens, gp63 and LACK have been used as primed boost vaccination by this strategy. In another experiment the same regimen had been applied using KMP11 against L. donovani infection. Promising effects of such immunization process have been approved by activation of CD4 and CD8 specific responses with higher levels of IL-2 , IFN-γ and TNF in vaccinated group [2]. Other advantages of this delivery system include its suitability to act as an attenuated recombinant vaccine and the capacity to have up to 30 Kbp heterologous DNA. However, there are some safety concerns such as potential cytotoxicity, high immunogenicity and transient expression of the transgene which need to be considered before administration [3]. Influenza virus is another viral delivery system which is used as an anti-leishmanial vaccine vehicle for expressing LACK 158–173 antigen [4]. Similar concerns as Vaccinia delivery system should be considered regarding their administration. One of the most interesting experiments in viral delivery systems involves the application of attenuated canine distemper virus (CDV) as a system for delivering Leishmania antigens. In this study, three different Leishmania antigens (TSA, LACK, and LmSTI1) were delivered individually or as a cocktail in dog model against L. major infection. Although rCDV-LACK could reduce the wound size more effectively than TSA and LmSTI1 alone, but the best results achieved by using a combination of three antigens [5] (Table 1). Bacterial delivery vehicles: Bacterial delivery systems are mainly based on two different strategies, (i) application of bacteria with tendency to gastrointestinal tracts like Lactococcus lactis, Salmonella typhi, and Listeria monocytogenese, which are usually administered orally (ii) application of Bacillus Calmet Guerin (BCG) as a strong stimulator of cellular response in the body which is administered subcutaneously. Among the oral vaccine candidates, Lactococcus lactis is a safe non colonizing bacteria which is used in dairy products and categorized as GRAS (Generally recognized as safe) and therefore is planned for immuno-compromized patients as an oral route of administration. The room temperature stability makes it an ideal option for remote areas in underdeveloped countries. Hugentobler et al. co-expressed LACK and mouse IL-12 in Lactococcus lactis and showed that the vaccine can reduce lesion size in cutaneous infection via oral or subcutaneous administration [6,7]. In another experiment, Lactococcus lactis harboring A2 antigen was tested

against VL through sub-cutaneous route of administration and it was reported to be capable of reducing the parasite burden in liver [8]. Listeria monocytogenes is another carrier candidate with the ability of colonization within the mesenteric lymph nodes. As a sand fly transmitted disease, salivary gland proteins have crucial effect on disease outcome and act as potential vaccine candidates. Abdallah et al. utilized Listeria monocytogenes as a vehicle to carry LJM11, an antigen of Lutzomiya longipalpis sand fly salivary gland against L. major infection in C57BL/6 mice. The vaccinated group exhibited high levels of CD4+/IFN-γ+/ TNF-α -/IL-10- or CD4+/IFN-γ+/ TNF-α +/IL-10- cells. Smaller lesions and lower parasite load was also observed in the vaccinated group [9].

Downloaded by [] at 03:00 01 March 2016

Salmonella typhimurium as an intracellular bacterium which creates systemic infection can stimulate strong humoral, cellular and mucosal responses. The ultimate destination of Salmonella typhimurium is the phagosomes inside monocyte-derived cells. This property can make the attenuated form of Salmonella. typhimurium an ideal vehicle as a vaccination strategy against leishmaniasis for delivering antigen of interest into APCs (Antigen presenting cells) [10]. Schroeder et al. administered recombinant Salmonella typhi expressing 5 different antigens in BALB/c mice against L. major and L. donovani infection. According to the results, two of the antigens (LinJ08.1190 and LinJ23.0410) resulted in more reductions in L. major visceralization and improvement in the resistance against L. donovani infection, compared to the other ones [11]. BCG as one of the earliest vital vaccines has been successfully applied to immunize the world population against mycobacterium tuberculosis. BCG spreads from the inoculation site to the lymphatic tissue and stimulates cellular immune responses. In natural form of mycobacterium tuberculosis infection, the immunity is elicited through the activation of macrophages with sensitized T cells (http://apps.who.int/iris/bitstream/10665/44733/1/9789241502412_eng.pdf). Therefore, the mechanism of action is somehow similar to Leishmania infection so it has the potential to act as an ideal vector for Leishmania antigens to simulate the cellular immune responses. BCG has been used as preventive vaccine against L. tropica infection in BALB/c mice. It reduced the severity of the disease, inhibited from visceralization and significantly increased the survival chance [12]. BCG expressing gp63 has been used to immunize BALB/c and CBA mouse against L. major and L. mexicana infections. Both mice could significantly resolve L. mexicana infection. Infection in CBA was controlled against L. major but BALB/c mice only showed a delay in lesion formation [13]. In another study, Abdelhak. S. et al. investigated the protective response of two different recombinant BCG expressing gp63 in C57BL/6 and BALB/c mice. They observed that the Recombinant BCG producing gp63 as a hybrid protein with the N-terminal region of the β-lactamase can induce protective response against L. major in BALB/c mice [14]. Streit J.A. et al. vaccinated BALB/c mice with BCG expressing L. chagasi LCR1 antigen through different intraperitoneal (i.p.) and subcutaneous (s.c.) routes of injection. Only the subcutaneous rout could elicit protective immune response with increased IFN-γ and decreased IL-10 levels against L. chagasi challenge [15] (Table 2).

Parasitic delivery vehicles: Leishmanization

Downloaded by [] at 03:00 01 March 2016

The central dogma in vaccination process is that, the vaccine should be able to simulate sufficient immunological response to combat the infectious agent. Successful experiment on Smallpox vaccination which eradicates the disease is an excellent example. In such study, the cowpox virus was used for live vaccination against smallpox in human. From ancient times, people in endemic areas traditionally used exudates of Leishmanial active lesions to inoculate children's arms to prevent from disfiguration in the face and other exposed part of the body [16] called as Leishmanization. Actually, leishmanization is the intentional infection with inoculation of standard count of parasite to induce active immunity against Leishmania infected sand fly bites. This approach has been practiced for a while in the former Soviet Union and Middle East before and during the gulf war. Although this approach has been considered as the best practical way of vaccination against cutaneous form of the disease until now, it has been stopped according to safety concerns and unusual immunological responses of some sensitive individuals [17].

Live attenuated parasites Being useful for eliciting lifelong protective responses, live Leishmania vaccines have attracted a great deal of scientific interest with the aim of putting an end to this ancient health problem. Many attempts have been made to introduce a live Leishmania vaccine which can meet the safety requirements and also elicit adequate immunity to prevent the occurrence of disease. Live attenuated Leishmania vaccines are being explored as potential candidates. Safety of the attenuated parasites in terms of non-pathogenicity, host survival limitation, and non-revision to virulence are important aspects of the safety profile of these vaccines. Furthermore, gene selection for the knock out process is a crucial issue to limit their pathogenicity and preserve the immunogenicity. Various strategies have been tested to create live vaccines against leishmaniasis, including (i) knocking out essential genes in virulent parasite (ii) transfection of non-pathogenic vectors like Leishmania tarentolae with immunogenic antigen sequences (iii) construction of suicidal parasites to prevent pathogenicity (iv) combination of live parasites with CpGoligodeoxynucleotide (CpG ODNs) motifs.

Knocking out essential genes in virulent parasite as a vaccination tool Dey et al. attenuated L. donovani parasite by knocking out two genes, LdCen and LdP27 and tested it against infectious challenge with L. mexicana in BALB/c mice. This regimen induced strong T cell response with down regulation of Th2 cytokine profiles [18].

LdCen deficient L. donovani parasite has also been tested against L. infantum infection in dogs, resulting in reduced parasite load by 87% after 18 months post challenge [19]. Furthermore, Selvapendiyan et al. have shown that LdCen1-/- can partially protect BALB/c mice, hamster and SCID mice against L. donovani and L. braziliensis challenge by eliciting IFN-γ, IL-2 and TNF-α production [20]. In another work, Cysteine proteinase (cp) deficient mutants of L. mexicana parasites vaccine was used and demonstrated to decrease Th2 response against L. meixcana infection in hamster model [21].

Downloaded by [] at 03:00 01 March 2016

Carrion et al. studied the HSP70-II L. infantum null mutant vaccine against L. major infection in BALB/c mice. They compared the efficacy of the vaccine in three different routes of injections namely intravenous (i.v.), intraperitoneal (i.p.) and subcutaneous (s.c.). Furthermore, the construct was safe for immunodeficient SCID mice [22]. One of the major genes in Ascorbate biosynthesis pathway is Arabino-1, 4-lactone oxidase (ALO) enzyme in L. donovani. Anand et al. studied the effect of null mutants of ALO against L. donovani challenge in BALB/c mice. They found that the mutant parasite is safe and induce protective immunity against L. donovani [23].

Construction of suicidal parasites to prevent pathogenicity as a vaccination tool Some other experiments tried to control the vaccinated parasite using exogenous processes such as photodynamic or drug sensitive suicidal mutants. In one study, Leishmania was transformed with 2 different mammalian genes of heme biosynthesis pathway, delta-amino levulinate (ALA) dehydratase and porphobilinogen deaminase. These genes are originally absent in Leishmaina and cause sensitivity to UV irradiation. After vaccination with the construct, ALA was exogenously given and the enzymes changed the Aporphyria to Uroporphyria. The Porphyrin which was excited by light to produce leishmanolytic oxidative species destroyed the live parasite. Photodynamic vaccination of hamsters with the suicidal mutants decreased the parasite load by 99% and suppressed the development of disease [24,25]. Davoudi et al. constructed a drug sensitive L. major parasite which was harboring Thymidin kinase (tk) and Cytosine deaminase (cd) genes. The former gene increased the sensitivity to gancyclovir and the second one sensitized the parasite to 5fluorocytosis (5-FC). BALB/c mice lesions were cured in 2 weeks following treatment with drugs either alone or in combination. In addition to this, in a similar experiment C57BL/6 mice exhibited a strong Th1 response [26].

Recombinant nonpathogenic L. tarentolae as a vaccination tool Recently, a nonpathogenic Leishmania parasite has been isolated from a lizard named L. tarentolae which is unable to activate infection in human. Different approaches have been

exploited to change the parasite into a vehicle for delivering antigens of interest to the immune system. For the first time, Breton et al. showed that vaccination with L. tarentolae can protects BALB/c mice against L. donovani challenge [27].

Downloaded by [] at 03:00 01 March 2016

A2 gene which is believed to be responsible for viscerotropic nature of L. donovani and L. infantum, was transfected into the L. tarentolae and used as a vehicle vaccine against L. infantum infection in BALB/c mice. Protective response was associated with high levels of IFN-γ and reduced levels of IL-5 [28]. Some other studies have been dedicated to test Live and DNA vaccinations alone or in combination as a potent approach to immunize mice. In our recent work, recombinant L. tarentolae harboring CPA/CPB along with DNA vaccine using PpSP15 was applied in C57BL/6 and BALB/c mice models. The best results were obtained with DNA priming with PpSP15 followed by live recombinant L. tarentolae expressing CPA/CPB as booster regimen [29]. In another attempt, a recombinant L. tarentolae expressing the tri-fused genes A2-CPA-CPBCTE (CPB without C terminal) was produced as a new live vaccination strategy against visceral Leishmaniasis. Two modalities namely DNA/live and Live/live vaccinations were performed in BALB/c mice against L. infantum infection. We showed that immunization with prime-boost DNA/live strategy elicited promising immunization against high dose of L. infantum challenge [30]. Furthermore, we vaccinated outbred dogs with a prime-boost regimen based on recombinant L. tarentolae expressing the tri-fused gene A2-CPA-CPB and evaluated its immunogenicity and protective immunity against L. infantum infectious challenge. Based on the results, vaccinated animals produced partial protection with significantly higher levels of IgG2, (but not IgG1), and also IFN-γ and TNF-α, but low IL-10 levels, before and after challenge as compared to control animals [31]. Recently, we have generated a recombinant non-pathogenic L. tarentolae-PpSP15 parasite and tested it in the presence of CpG ODNs as a novel strategy against L. major infection in BALB/c mice. We observed high levels of IFN-γ and IL-17 production pre- and also post-challenge against L. major in susceptible BALB/c mice. This is the first report showing the effect and applicability of live nonpathogenic Leishmania secreting a sand fly salivary protein in the presence of CpG ODNs [32] (Table3).

Chemical Leishmania antigen delivery systems Liposomes as Leishmania antigen delivery systems Liposomes are composed of natural amphiphilic phospholipids which are nontoxic and nonimmunogenic. Within such phospholipids, whether the antigen is lipophilic or hydrophilic, it can be found in the lipid layer or inside the aqueous core, respectively. In 1974, liposomes were applied as immunological adjuvants for the first time. Since that time, liposomes have been frequently used as suitable antigen delivery vehicles for slow releasing and depository effects in vaccine studies. Liposomes have been considered as antigen delivery systems, as they are

Downloaded by [] at 03:00 01 March 2016

able to incorporate a wide variety of antigens, such as malaria, hepatitis A, tetanus toxoid, diphtheria, influenza and leishmanial proteins, and can be accessed with various compositions, bilayer fluidities, sizes and charges [33]. The most important functions of liposomes in vaccine-related applications include the protection of antigens against being cleared from the body and the delivery of such antigens to the professional antigen-presenting cells (APC). Liposomes can facilitate antigen migration and delivery into the APC cytosol to induce humoral as well as cell-mediated immune (CMI) responses, under the pathways of antigen presentation by MHC class I and II. It has been suggested that liposome uptake occurs via phagocytosis, endocytosis or pinocytosis processes [34]. Liposomes are also able to increase the expression of various chemokine genes, such as CCL2, CCL3, and CCL4 by dendritic cells. In addition, liposomes act to protect antigens from fast degradation inside the APC and hence prolong in vivo T cells primary activation [35]. Application of liposomes as vaccine delivery systems is associated with some advantages such as their easy preparation, biodegradability and low toxicity. However, Their efficacy is affected by various physical factors such as their size, surface charge, phospholipid composition, and vesicles stability [36]. In the following, some examples about several parameters of liposomes evaluated as antiLeishmania antigen delivery systems are discussed. In terms of liposome size, in a previous study, it has been demonstrated that the balance between Th1/Th2 responses can be altered by changing the size of vesicle. In this work, after preparing lipid vesicles with three sizes of 100, 400 or 1000 nm encapsulating rgp63, the effect of vesicle size on the immune response type as well as protection against leishmaniasis in BALB/c mice was investigated. According to the results, making use of larger liposome sizes (400 and 1000 nm) resulted in smaller footpad lesions and parasite burden in the spleen of BALB/c mice after the challenge infection with L. major. In addition to this, the production of IFNγ by spleen cells was only increased in the mice immunized with larger liposomes (400 and 1000 nm sizes). This research demonstrated the crucial role of liposome size in determining the type of immune response. In murine model, larger liposomes (with sizes equal or greater than 400 nm) induce CMI response, while smaller liposomes (100 nm size) promote humoral immune response against leishmaniasis [37]. The surface charge of a vesicle has been reported to significantly influence the adjuvant effect of that liposome. In fact, the net surface charge of liposomes could be changed by combining positively charged (e.g. DDAB, stearylamine, dioleoyl trimethyl ammonium propane) or negatively charged lipids (e.g. phosphatidic acid, phosphatidyl glycerol, phosphatidyl serine (PS) or DCP). For instance, Afrin et al. prepared negatively and positively charged liposomes using phosphatidic acid and stearylamine, respectively. They found that in comparison with the negatively charged liposome (consisting of egg lecithin/ phosphatidic acid/ cholesterol; 7:2:2) or the neutral one (consisting of egg lecithin/ cholesterol; 7:2), the positively charged liposome (consisting of egg lecithin/ stearylamine/ cholesterol; 7:2:2) encapsulating L. donovani promastigote membrane antigens (LAg) resulted in a considerable protection against visceral leishmaniasis, with delayed-type hypersensitivity and antibody responses [38]. In another study, the adjuvanticity and also the protective efficacy of positively charged liposomes (consisting of egg lecithin/ stearylamine/ cholesterol; 7:2:2) against L. donovani in BALB/c mice were found to be higher than those of the negatively charged liposomes

Downloaded by [] at 03:00 01 March 2016

(consisting of egg lecithin/ phosphatidic acid/ cholesterol; 7:2:2) or the neutral ones (consisting of egg lecithin/ cholesterol; 7:2) [39]. Immunization using soluble Leishmania antigen (SLA) alone and in positive, neutral and negative liposomes resulted in different protection levels, correlated with IL-4 and IFN-γ production, in which for maximum protection, there was a skewing toward IFN-γ producing Th1-type immune response. It was also demonstrated that SLA in positively charged liposome vaccine, could be applied for immunotherapy in murine against established visceral infection. The results showed that desirable resistance was associated with stimulation of Th1-type immune responses and inhibition of IL-4 and IL-10 production [39]. In the study performed on BALB/c mice by Badiee et al., to assess the effect of liposome charge on stimulating a Th1-type immune response and protection against leishmaniasis, negatively and positively charged liposomes were produced by the addition of DCP or DDAB to the neutral liposome, respectively. They found that in comparison with positively charged liposomes (consisting of DPPC/ DDAB/ cholesterol; 2:1:1), incorporation of recombinant gp63 in neutral liposomes (consisting of DPPC/ cholesterol; 2:1) is more effective in promoting Th1-type immune response, while using rgp63 in association with negatively charged liposomes (consisting of DPPC/ DCP / cholesterol; 2:1:1) resulted in a Th2- type immune response [40]. Hence, such studies are demonstrative of the essential role of surface charge on the extent and type of the resulting immune response. Regarding the phospholipid composition of the liposome, in a study by Bhowmick et al., the vaccine potential corresponding to leishmanial antigen (LAg), making use of reverse-phase evaporation vesicles (REV), dehydration–rehydration vesicles (DRV), or multilamellar vesicles (MLV) to protect BALB/c mice against experimental VL was assessed [41]. While a partial resistance was observed in the case of LAg alone or incorporated in REV, nearly complete protection occurred by using cationic liposomes prepared by both MLV and DRV methods. Such protection was mainly of Th1-type immune responses, indicated by the increase in DTH and IgG2a isotype antibodies as well as IFN-γ production. Encapsulation of LAg in MLV was associated with durable CMI and it was observed that mice challenged for ten weeks following vaccination were also able to strongly resist the experimental challenge [41]. In order to improve the vesicle stability, one can replace under vitalized phosphatidylcholine (PC, Tc −10 ◦C) with phospholipids associated with high transition temperatures (Tc) [42] like Distearoyl phosphatidylcholine (DSPC, Tc 54 ◦C), Dipalmitoyl phosphatidylcholine (DPPC, Tc 41 ◦C) and dimyristoyl phosphatidylcholine (DMPC, Tc 23 ◦C). In the case of membranes containing antigens with high molecular mass, a correlation between the Tc of phospholipids and the resulting immune response (cellular or humoral) has been reported [43]. An optimal fluidity of liposomes seems to be required for them to be captured by APCs. In one of previous studies by Badiee et al., after preparation of liposomal rgp63 associated with different phospholipids (EPC, DPPC or DSPC), the corresponding adjuvanticity of such liposomes against challenge with L. major in BALB/c mice were compared. They found that the liposome bilayer composition affects the resultant immune response in mice such that immunization with liposomes associated with DPPC or DSPC promoted Th1-type immune response while the ones with EPC led to a Th2-type immune response [44]. Furthermore, in another work, LAg encapsulated in lipid vesicles which had been produced utilizing cholesterol and DSPC (Tc= 54 ◦C), were used for intraperitoneal immunization.

Downloaded by [] at 03:00 01 March 2016

According to the results, protection against L. donovani challenge was associated with differentiation of IFN-γ producing Th1 cells and DTH responses [45]. These results showed that liposomes which are prepared with higher Tc phospholipids seem to be more efficient in promoting the Th1-type immune response and protection, and hence can be further investigated to produce an optimized vaccine against leishmaniasis. In the work carried out by Shimizu et al. on BALB/c mice, SLA encapsulated in liposomes which were coated with neoglycolipids containing oligomannose residues (Man3 or Man5) was intraperitoneally administrated and the results were indicative of strong induction of an antigenspecific Th1-type immune response [46,47]. In such studies, induction of the protective response in BALB/c mice against L. major infection may be due to the intense induction of a Th1-type immune response specific to the encapsulated antigen, after immunization by intraperitoneal administration of SLAoligomannose-liposome, and its stimulation is also triggered by peritoneal CD11b+ cells, i.e. macrophages. In another study, the effect of sphingomyelin (SM) liposomes associated with SLA on the promoted immune response against leishmaniasis was examined. The SM-liposome-SLA promoted strong Th2-type immune responses, so cannot be considered as a suitable strategy to promote Th1-type immune response and protect the BALB/c mice against Leishmania infection [48]. Furthermore, Sharma et al. produced non-phosphatidylcholine (non-PC) liposomes, i.e. escheriosomes from E. coli lipids and observed that in comparison with sLAg entrapped in egg PC/ cholesterol liposome (EPC-sLAg) or sLAg performed with incomplete Freund’s adjuvant. Immunization of BALB/c mice through intraperitoneal administration of escheriosome encapsulated sLAg (EL-sLAg) induced a more intense CD8+ cytotoxic T lymphocyte (CTL) response. In another set of experiments, hamsters immunized with EL-sLAg were better protected compared to the EPC-sLAg-immunized hamsters. They speculated that escheriosomes can be used to deliver the desired antigen to cytosol of APCs [49]. To evaluate the protective impacts of liposomes of Leishmania antigens for inducing the immune system against leishmaniasis, different routes of immunization have been applied. In a number of studies, different liposome encapsulated Leishmania antigens such as rgp63 [37,40,44,50-52], sLAg [53] and rLmSTI1 [54,55], have been applied for subcutaneous route. Other works have focused on the intraperitoneal administration of liposome entrapped antigens such as L. donovani promastigote membrane antigens, LAg [38,41,45,56-59], L. major lipophosphoglycan, LPG [42], L. donovani promastigote soluble antigens, sLAg [39,46,47,49,60], gp63 derived from L. donovani promastigotes [61], and some polypeptides isolated from L. donovani [62], which have been reported to promote protection against leishmaniasis in BALB/c or Hamster animal models, as summarized in table 4. With the purpose of understanding the immunological correlation between the protective and non-protective routes, Bhowmick et al. made a comparison between the protective efficacy corresponding to LAg either free or incorporated in positively charged liposomes through four different administration methods, namely i.v., i.p., s.c. and intramuscular (i.m.) routes against L. donovani infection in BALB/c mice. It was observed that compared to mice immunized via the s.c. and i.m. routes which were not protected, the mice immunized i.v. and i.p. using LAg, either

Downloaded by [] at 03:00 01 March 2016

alone or entrapped in liposomes, were protected against the challenge infection with L. donovani [63]. In another work by the same group, the protective efficacy in the BALB/c mice model was enhanced by making use of leishmanial antigen in association with Toll like receptor (TLR) agonists monophosphoryl lipid A-Trehalose dicorynomycolate (MPL-TDM) or cationic liposomes against experimental VL using the i.p. administration method [58]. Because of inadequacy of the intraperitoneal method for human immunization, in another research they utilized vaccines which were combinations of cationic liposomes and MPL-TDM, making use of rgp63 as the protein antigen through s.c. route. Such injections caused enhanced immune responses which further induced high level protection against VL in the studied mouse model [64]. They also assessed the immune response and protection promoted by liposome of SLA in association with MPL-TDM via subcutaneous administration and found that such BALB/c mice immunization using SLA encapsulated in liposomes or in association with MPL-TDM induced partial protective levels against experimental visceral leishmaniasis. However, adjuvanting liposomal SLA with MPL-TDM resulted in much higher protective levels in the spleen and liver of the mice infected with L. donovani [65]. Liposomal Combination is also a suitable vaccine delivery system and MPL-TDM as an immunopotentiator has been validated by such researches. Das and Ali also demonstrated the higher efficacy of an antigenic cocktail of type I, II and III CPs encapsulated in cationic liposomes in association with MPL-TDM in a hamster model against L. donovani [66]. The cysteine proteases of Leishmania encapsulated in cationic liposomes associated with MPL-TDM provide a desirable class of vaccines in human VL to be further studied. In a previous study, the ability of L. donovani (78 kDa antigen) by itself or in association with various adjuvants, namely autoclaved Leishmania donovani antigen (ALD), liposomal encapsulation, MPL-A, recombinant IL-12, and Freund’s adjuvant (FCA) against visceral leishmaniasis in murine model was investigated. According to the results, the vaccine consisting of 78 kDa antigen and rIL-12 exhibited the highest level of protective efficacy, after which the liposome incorporated 78 kDa and 78 kDa +MPL-A vaccines were observed to elicit Th1-type immune response indicated by the elevated IL-2 , IFN-𝛾, and IgG2a production [67]. In some works, co-encapsulation of liposomal antigens with other adjuvants like non-coding pDNA harboring CpG motif [60], CpG ODNs [51,53,55,68,69], or BCG [70] has resulted in the enhanced immune response and higher protection against leishmaniasis. The findings of above studies indicate that Leishmania antigens in co-incorporation with immunostimulatory adjuvants like MPL or CpG motif in cationic liposome are able to elicit protective immunity against leishmaniasis. Non-Ionic Surfactant Vesicles (Niosomes) as Leishmania antigen delivery systems Niosomes, non-ionic surfactant vesicles (NSVs) are prepared via hydrating synthetic mono- or di-alkyl surfactants along with proper amounts of cholesterol or other amphiphilic molecules. Niosomes, as either unilamellar or multilamellar are capable of encapsulating both hydrophilic and lipophilic materials like drugs or vaccines. They are also able to elicit both cell-mediated and humoral immune responses against a number of antigens such as bovine serum albumin and hepatitis B [71].

There are two studies regarding anti-Leishmania vaccine using niosome. LezamaDávila CM et al. demonstrated that subcutaneous administration of purified gp63 encapsulated in niosomes (but not liposomes) induced protective immune responses in C57BL/10 mice against cutaneous leishmaniasis, and only ulcerated lesions which started to heal, in comparison with the control mice which only showed unulcerated lesions with no indication of healing [72]. In another study, Pardakhty et al. reported that in contrast to the autoclaved L. major (ALM), the ALMcontaining niosomes are more able to prevent cutaneous leishmaniasis in BALB/c mice [73]. Niosomes exhibit some advantages over liposomes, such as their cost-effective production, high stability and purity, uniformity of their content, easy storage as well as easier handling, which make them desirable candidates for application in vesicle-system for releasing drug or antigen, and hence there is a need to further study the application and effects of niosome as antigen delivery system in Leishmania vaccine investigations.

Downloaded by [] at 03:00 01 March 2016

Virus like particles (Virosomes) as Leishmania antigen delivery systems Virus-like particles (VLPs) or virosomes which are structural proteins of viruses in the absence of genetic materials, are able to mimic the real virus appearance and carry the desired antigens on their surface with the purpose of eliciting an immune response against various microorganisms. Cervarix and Gardasil are two recent commercially available vaccines which have been successfully tested and approved by FDA for vaccinating against Human papillomavirus [74]. VLPs can be produced by cell expression systems in insect and mammalian bodies. Several viruses such as Hepatitis B and Parvovirus can be prepared as VLPs to carry the desired antigens on their surface [75]. So far only two experiments have been performed with virus like particles in Leishmania vaccine research. In one experiment, Zhou G et al. applied HbcAg (Hepatitis B core antigen) which had been joined to an IL-10 peptide to induce anti IL-10 polyclonal antibodies in mice. Although IL-10 restraining seems important for recovering from leishmaniasis, in this experiment, vaccinated group faced larger lesions and exacerbation of the disease [76]. In another study, Liu x et al. tried to test a carbohydrate base vaccine against visceral leishmaniasis. To elicit long term T cell related response, they conjugated synthetic Leishmania carbohydrates (LPG) to influenza virosomes (IRIVs) that induced both IgM and IgG isotype antibodies against Leishmania glycan in mice [77].

Cationic Solid Lipid Nanoparticles (cSLN) as Leishmania antigen delivery systems Cationic solid lipid nanoparticles (cSLN) are promising lipid Nano carriers used for different disciplines such as drug and vaccine delivery. These nanoparticles consist of physiologically well-tolerated ingredients mostly approved for pharmaceutical application in human [78]. The main characteristics of cSLN include their stability, no special requirement for packaging and their resistance to oxidation [79]. In comparison with liposomes, cSLNs have chemical stability and various surfanctants and lipids can be used in their design. cSLNs possess and act through

two options for delivering the antigen and acting as delivery system: i) encapsulation of antigen inside the lipid matrix of the SLN ii) absorption on the surface by electrical interaction with cationic SLNs. It is notable that beside chemical properties, physical parameters such as size, zeta potential and stability are also influential on the controlled release profile of target antigens.

Downloaded by [] at 03:00 01 March 2016

We have recently developed a novel SLN-based DNA and protein using cysteine proteinases as target antigen. In this regard, we have reported the formulation of immunogenic cpa and cpb genes with cSLNs consisting of cetyl palmitate, cholesterol, DOTAP and Tween 80, as a novel non-viral DNA vaccine formulation against leishmaniasis [80]. We demonstrated that stable formulation of cSLN was able to protect pDNA in DNase I challenge assay. In comparison with linear PEI-25kDa, the in vitro COS-7 cells transfection revealed that these formulations are associated with low cytotoxicity and acceptable efficiency/cytotoxicity ratio. Moreover, in another study we evaluated the potency of CPs types I, II and III combination formulated with cSLNs evaluated as DNA vaccination against L. major infection in BALB/c as susceptible mice model. In this study, the cSLN-pcDNA-cps was formulated into particles in nanometeric range (~240-250 nm) [81]. Beside the size parameter which facilitated the intracellular delivery of selected formulation , the presence of cholesterol domains and Tween 80 in cSLN enhanced the transfection efficiency and phagocytosis by APC as well as localization in draining lymph nodes following subcutaneous administration [82]. Furthermore, Doroud et al. investigated the protective efficacy of recombinant protein vaccine (CPB –CTE) formulated with SLN against L. major challenge in C57BL/6 and BALB/c mice. Promising effects of elevated level of IFN-γ and lower amounts of IL-4 as a Th2 cytokine were observed [83,84]. In another approach, Saljoughian et al., compared physical (electroporation) and chemical (cSLN formulation) delivery systems in the inbred mice model. In this study, a DNA vaccine harboring the L. donovani A2 antigen along with L. infantum cysteine proteinases were tested against L. infantum challenge in BALB/c mice model. The results showed that prime-boost administration of the pcDNA-A2-CPA-CPB-CTE delivered by either electroporation or cSLN formulation protects BALB/c mice against L. infantum challenge. The protective immunity is associated with high levels of IFN-γ and lower levels of IL-10 production and induced strong Th1 immune response. Therefore, it was demonstrated that cSLNs as a nanoscale vehicle of Leishmania antigens can improve immune response and act as a promising strategy against visceral leishmaniasis [85]. Following this finding, similar approaches were applied in canine leishmaniasis and both electroporation and cSLN were utilized as delivery system. The observed partial protection in vaccinated dogs is associated with significantly higher levels of IgG2, IFN-γ, and TNF-α and with low levels of IgG1 and IL-10 as compared to the control group. The obtained protection was correlated with a low parasite burden and a strong DTH response in vaccinated animal using two delivery system without any significant differences [86].

Alginate as Leishmania antigen delivery systems

Downloaded by [] at 03:00 01 March 2016

Alginate is a water-soluble and linear natural polysaccharide consisting of alternating blocks of 1–4 linked α-L-guluronic and β-D-mannuronic acid residues. It can be readily cross-linked into a solid matrix by adding di- or tri-valent cations. Because of the safety, biodegradability and also being a low-cost polymer, alginate seems as a promising choice for application in vaccine delivery. Recent studies on this substance are indicative of its potential for application in immunoadjuvant and vaccine delivery system [87]. Utilization of alginate microspheres containing antigenic proteins for animal Immunization induced both cell-mediated and humoral immune responses. Tafaghodi et al. has optimized the production method of alginate microspheres incorporated with L. major (ALM) and QS adjuvant [88] and demonstrated that subcutaneous immunization of BALB/c mice with ALM encapsulated in alginate microspheres, ALM with CpG ODNs, and ALM co-encapsulated with CpG ODNs in alginate microspheres induced CMI and protected mice against L. major infection. Mice administration of alginate microspheres co-incorporated with CpG and ALM exhibited the significantly highest protection level [89]. Poly methyl methacrylate (PMMA) as Leishmania antigen delivery systems Poly methyl methacrylate (PMMA) is a FDA-approved synthetic polymer for specific human clinical applications such as bone cement. In vivo, the PMMA particles are phagocytosable , and can induce intense immune responses via eliciting inflammatory cytokines production [90]. Such nanoparticles can be easily prepared with the desirable particle sizes and surface properties, and among their numerous advantages one can name their simple and easy production, low degradation and mild adverse effects during usage. In one experiment, Zarrati et al. reported PMMA to affect the efficacy of a DNA vaccine encoding TSA in BALB/c mice against L. major infection, inducing humoral immune responses to the delivered antigen [91]. Poly Lactic-co-Glycolic Acid (PLGA) as Leishmania antigen delivery systems PLGA, poly lactic-co-glycolic acid, is a well-known biodegradable and biocompatible polymer that can be prepared as nano- or microparticles, which are promising protein, peptide and DNA vaccine delivery systems. PLGA has been long approved for use in human body in controlledrelease delivery systems for some drugs [13,92]. In fact, PLGA is a lactic and glycolic acid copolymer, and is broken down into its monomers, which are natural metabolites and biocompatible. PLGA microparticles incorporating antigen can enhance cross-presentation and elicit CTL responses [93]. In a study by Tafaghodi et al., it was demonstrated that when ALM encapsulated in PLGA nanospheres (mean diameter of 294 ± 106 nm) can elicit intense Th1 protective immune response in BALB/c mice model. On the contrary, Quillaja saponins adjuvant exhibited a reverse effect and resulted in the least protective responses [94]. In another study in BALB/c mice, they demonstrated that immunization by making use of CpG and PLGA nanospheres incorporated with ALM caused reduced L. major infection in association with reduced IL-4 and enhanced IFN-γ production [95]. Santos et al. has reported that priming with PLGA nanoparticles incorporated with DNA encoding L. infantum chagasi KMP-11 and then PLGA nanoparticles incorporated with

Downloaded by [] at 03:00 01 March 2016

recombinant KMP-11 elicits higher levels of protective cellular immune responses in BALB/c mice, in comparison with immunization using DNA encoding L. infantum chagasi KMP-11 against L. braziliensis plus sand fly saliva [96]. Sofia A Costa Lima reported that PLGA nanoparticles provide controlled and effective delivery of Bisnaphthalimidopropyldiaaminooctane (BNIPDaoct) for treatment of VL [97].

Immune Stimulating Complexes (ISCOMs) and ISCOMATRIX as Leishmania antigen delivery systems ISCOMs can be prepared by phospholipids, cholesterol, envelope protein antigen and Quil A in Triton X-100 combined with a variety of antigens, while ISCOMATRIX is an adjuvant that can be prepared by phospholipid, cholesterol and saponin without any antigen. ISCOMATRIX vaccines (ISCOMATRIX adjuvant combined with antigen) have been recently tested in clinical trials and demonstrated to be safe and able to induce intense antigen-specific cellular or humoral immune responses against a wide range of antigens. Sjo¨lander et al. demonstrated that vaccination of mice with PSA-2 DNA results in protection against L. major infection, and the mice exhibits a virtually exclusive Th1-type immune response. On the contrary, PSA-2 ISCOMs (consisting of phosphatidylcholine/ ISCOPREP 703 (Quil A )/ cholesterol; 1:3:1) elicit a mixed immune response (of Th1- and Th2-types), being unable to provide protection [98]. C3H/He mice vaccination with native promastigote PSA-2 and C. parvurn as the adjuvant elicits a Thl-type immune response protecting the mice against infection. It was found that while PSA-2 purified from E. coli and administered in ISCOMs or with C. parvum as adjuvant induces intense Thl-type responses, is unable to elicit protection [99]. Papadopoulou et al. have reported that intraperitoneal administration to vaccinate BALB/c mice with low doses of gp63 into ISCOMs promotes partial protection against L. major which directs the immune response towards a Th1- rather than Th2-type [100]. In another study, it has been suggested that positively (consisting of DOTAP/Quil A/cholesterol; 2:2:1) or negatively (consisting of DSPC/Quil A/cholesterol; 2:2:1) charged ISCOMATRIX plus SLA elicit a mixed immune response (of Th1- and Th2-types) in BALB/c mice and is not protective against L. major [101]. Stable nano-emulsion formulations: Stable emulsion formulations can be used as vaccine adjuvants in combination with recombinant protein candidates. MPL (Manophosphoryl lipid A) and its synthetic homologue, GLA (Glucopyranosyl lipid A) are TLR4 agonists which can induce Th1 response against infections and cancer. GLA is able to activate cytotoxic activity of CD4+ T cells through mechanisms independent of Fas ligand-Fas, TRAIL-DR5 and canonical death pathways. The molecule can induce Granzyme A expression through an unknown mechanism which requires the expression of CD145 (CD40L) in T cell and CD40 in the target cell. GLA increases IFN-γ and TNF-α T cell response in oil-in-water emulsion [102]. In a recent experiment Coler RN et al. formulated LEISH-F3 polyprotein of Leishmania in a stable oil-in-water nanoemulsion (SE) using GLA-SE. According to their results, the formulation was able to induce protection in vaccinated groups against L. donovani and L. infantum

challenges in mice. The mentioned vaccine, which is the only vaccine candidate in the first phase of human clinical trials, has been tested in healthy volunteers in USA and its safety and efficacy has been approved based on the protective cytokine and Immunoglobulin production [1].

Downloaded by [] at 03:00 01 March 2016

NS Daifalla et al. formulated two different L. donovani recombinant antigens, namely iron superoxide dismutase B1 (SODB1) and Peroxidoxin4 (Pxn4) with GLA-SE. The vaccine could induce Th1 response through induction of higher levels of IFN-γ and lower levels of IL-10 production [103]. Furthermore in another experiment using the same formulation, reduced parasite load and increased level of Th1 cytokines could be observed in the challenged footpad of vaccinated group, compared to the control group [104]. R Gomes et al. also tested GLA in formulation with two different recombinant protein antigens (KSAC and L110F), which were both formulated with GLA, L110F also formulated with MLP. In this research needle challenged groups were compared to the one challenged with sand fly bites. In the needle challenged vaccinated groups both formulations were protective while in natural challenged groups, only KSAC-GLA was able to induce protective immune response [102]. In another study, such antigens were formulated with GLA, and tested against L. major challenge in C57BL/6 mice. Vaccinated animals exhibited a 60 fold increase in the number of CD4+ IFN-γ+ T cells compared to controls. They also exhibited a 100 fold decrease in the parasite load. Re-challenging vaccinated group with infected sand fly bites induced higher number of neutrophils in the challenged site and reduced the number of CD4+ IFN-γ+:IL-17+ T cells in comparison with controls [105].

Expert commentary and five year view Delivery systems have attracted a great deal of interest for vaccine development and are popular these days due to the benefits they offer. For subunit vaccines, it is highly desirable to apply the combination of delivery systems, immunopotentiators and isolated antigens for elicitation of the required immune response against parasite. Such combinations appear to stimulate both innate and adaptive immune systems. Live attenuated as well as recombinant non-pathogenic parasites can also broaden the scope for vaccine development. Safety of the attenuated parasites in terms of non-pathogenicity, survival limitation in the host, and nonrevision to virulence are important issues to be considered. Transfection of non-pathogenic parasites like L. tarentolae with salivary gland sequences and immunopotentiators such as CpG-oligodeoxynucleotide motifs are very exciting subjects to be exploited. The efficacy of these types of trials should be first applied in larger animals such as dog. Although, we need more investigation and regulatory confirmation, instead of using live Leishmania, the so called Leishmanization, we could replace it with non-pathogenic recombinant L. tarentolae.

Key issues:

• • • • • • •

Downloaded by [] at 03:00 01 March 2016



Vaccination is an approach to prevent leishmaniasis and until now there is no approved vaccine and only one is under evaluation in clinical trials for human. Different antigens have been found to induce protective immunity against Leishmania infection, but the major obstacle is their short term immunity. Delivery systems can maximize the rate or the extent of antigen effect. Delivery systems that can target monocyte/macrophages might potentially open up a new window for vaccine development against leishmaniasis. Intracellular microbes not only mimic Leishmania natural infection but also can play roles as carriers of special antigens. Non-pathogenic L. tarentolae can be a promising vaccine carrier that remains enough in the tissue. Various chemical carriers such as Liposomes, Niosomes, Virosomes, SLN, Alginate, GLA-SE and PLGA have been applied as delivery systems to enhance the immune response against Leishmanial infections. Electrostatic interactions between positively charged particles (such as cationic liposomes or cSLN) and negatively charged cell surface of APC help particle binding and internalization of loaded antigen and can subsequently improve the induction of immune responses even using lower dose of antigen.

Financial and competing interests disclosure The authors are supported by the Pasteur Institute of Iran and the National Science Foundation of Iran. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Abbreviation: 5-FC: 5-fluorocytosis; ALA: delta-aminolevulinate; ALD: autoclaved Leishmania donovani antigen; ALM: autoclaved L. major; ALO: Arabino-1, 4-lactone oxidase; APC: AntigenPresenting Cells; BCG: Bacillus Calmet Guerin; cd: Cytosine deaminase; CL: Cutaneous Leishmaniasis; CMI: Cell-Mediated Immune; cp: Cysteine proteinase; CpG ODNs: cytosine– phosphorothioate–guanine oligodeoxynucleotide; cSLN: Cationic Solid Lipid Nanoparticles; DMPC: dimyristoyl phosphatidylcholine; DOTAP : 1,2-dioleoyl-3-trimethylammonium-propane; DPPC: Dipalmitoyl phosphatidylcholine; DRV: Dehydration–Rehydration Vesicles; DSPC: Distearoyl phosphatidylcholine; FCA: Freund’s adjuvant; GLA: glucopyranosyl lipid adjuvant; GRAS: Generally recognized as safe; HSP: heat shock protein; ISCOMs: Immune Stimulating Complexes; i.v.: Intravenously; i.p.: Intraperitoneally; i.m.: Intramuscular; KMP-11: kinetoplastid membrane protein-11; L. major: Leishmania major; LACK: Leishmania homologue of activated C kinase; LAg: L. donovani promastigote membrane antigens; LCR1: antigen cloned from

amastigote L. chagasi library; MC: Mucocutaneous Leishmaniasis; MLV: Multi-Lamellar Vesicles; MPL: Manophosphoryl lipid A; MPL-TDM: Monophosphoryl lipid A-trehalose dicorynomycolate; MPL-A: Monophosphoryl lipid A; PC: phosphatidylcholine; PLGA: poly(lacticco-glycolic acid); PMMA: Poly methyl methacrylate; PpSP15: Phlebotomus papatasi salivary protein15; PSA-2: L. major parasite surface Ag-2; rgp63: recombinant major surface glycoprotein of Leishmania; rLmSTI1: Recombinant L. major stress-inducible protein 1; REV: Reverse-phase Evaporation Vesicles; s.c.: Subcutaneous; SE: stable oil-in-water nanoemulsion; SGS: salivary gland sonicate; SM: sphingomyelin; SLA: Soluble Leishmania Antigens; TLR: Toll Like Receptor; Tc: transition temperature; tk: Thymidin kinase; TSA: Thiol-Specific Antioxidant Protein; VL: Visceral Leishmaniasis; VLP: virus-like particle.

References:

Downloaded by [] at 03:00 01 March 2016

Reference annotations * Of interest ** Of considerable interest

1.

** 2.

3. 4.

5.

6.

7.

8.

Coler RN, Duthie MS, Hofmeyer KA et al. From mouse to man: safety, immunogenicity and efficacy of a candidate leishmaniasis vaccine LEISH-F3+ GLA-SE. Clinical & translational immunology, 4(4), e35 (2015). This is the only clinical trial on human vaccine against visceral leishmaniasis which had promissing results. Guha R, Das S, Ghosh J et al. Heterologous priming–boosting with DNA and vaccinia virus expressing kinetoplastid membrane protein-11 induces potent cellular immune response and confers protection against infection with antimony resistant and sensitive strains of Leishmania (Leishmania) donovani. Vaccine, 31(15), 1905-1915 (2013). Vannucci L, Lai M, Chiuppesi F et al. Viral vectors: a look back and ahead on gene transfer technology. New Microbiol, 36(1), 1-22 (2013). Kedzierska K, Curtis JM, Valkenburg SA et al. Induction of protective CD4+ T cellmediated immunity by a Leishmania peptide delivered in recombinant influenza viruses. PloS one, 7(3), e33161 (2012). Miura R, Kooriyama T, Yoneda M et al. Efficacy of Recombinant Canine Distemper Virus Expressing Leishmania Antigen against Leishmania Challenge in Dogs. PLoS Negl Trop Dis, 9(7), e0003914 (2015). Hugentobler F, Di Roberto RB, Gillard J et al. Oral immunization using live Lactococcus lactis co-expressing LACK and IL-12 protects BALB/c mice against Leishmania major infection. Vaccine, 30(39), 5726-5732 (2012). Hugentobler F, Yam KK, Gillard J et al. Immunization against Leishmania major infection using LACK-and IL-12-expressing Lactococcus lactis induces delay in footpad swelling. PloS one, 7(2), e30945 (2012). Yam KK, Hugentobler F, Pouliot P et al. Generation and evaluation of A2-expressing Lactococcus lactis live vaccines against Leishmania donovani in BALB/c mice. Journal of medical microbiology, 60(9), 1248-1260 (2011).

9.

10. 11.

12. 13.

14.

Downloaded by [] at 03:00 01 March 2016

15.

16.

* 17. 18.

19.

20.

21. 22.

23.

24. 25.

26.

Abdallah DSA, Bitar AP, Oliveira F et al. A Listeria monocytogenes-Based Vaccine That Secretes Sand Fly Salivary Protein LJM11 Confers Long-Term Protection against Vector-Transmitted Leishmania major. Infection and immunity, 82(7), 2736-2745 (2014). Roland KL, Brenneman KE. Salmonella as a vaccine delivery vehicle. Expert Rev Vaccines. 12(9): 1033–1045 (2013). Schroeder J, Brown N, Kaye P et al. Single dose novel Salmonella vaccine enhances resistance against visceralizing L. major and L. donovani infection in susceptible BALB/c mice. PLoS Negl Trop Dis, 5(12), e1406 (2011). Weintraub J, Weinbaum FI. The effect of BCG on experimental cutaneous leishmaniasis in mice. The Journal of Immunology, 118(6), 2288-2290 (1977). Connell ND, Medina-Acosta E, McMaster WR et al. Effective immunization against cutaneous leishmaniasis with recombinant bacille Calmette-Guerin expressing the Leishmania surface proteinase gp63. Proceedings of the National Academy of Sciences, 90(24), 11473-11477 (1993). Abdelhak S, Louzir H, Timm J et al. Recombinant BCG expressing the leishmania surface antigen Gp63 induces protective immunity against Leishmania major infection in BALB/c mice. Microbiology, 141(7), 1585-1592 (1995). Streit JA, Recker TJ, Donelson JE et al. BCG expressing LCR1 of Leishmania chagasi induces protective immunity in susceptible mice. Experimental parasitology, 94(1), 33-41 (2000). Khamesipour A, Dowlati Y, Asilian A et al. Leishmanization: use of an old method for evaluation of candidate vaccines against leishmaniasis. Vaccine, 23(28), 3642-3648 (2005). This article is the first repost for leishmanization in endemic area. Kumar R, Engwerda C. Vaccines to prevent leishmaniasis. Clinical & Translational Immunology, 3(3), e13 (2014). Dey R, Natarajan G, Bhattacharya P et al. Characterization of cross-protection by genetically modified live-attenuated Leishmania donovani parasites against Leishmania mexicana. The Journal of Immunology, 193(7), 3513-3527 (2014). Fiuza JA, Gannavaram S, da Costa Santiago H et al. Vaccination using live attenuated Leishmania donovani centrin deleted parasites induces protection in dogs against Leishmania infantum. Vaccine, 33(2), 280-288 (2015). Selvapandiyan A, Dey R, Nylen S et al. Intracellular replication-deficient Leishmania donovani induces long lasting protective immunity against visceral leishmaniasis. The Journal of Immunology, 183(3), 1813-1820 (2009). Silva-Almeida M, Pereira B, Ribeiro-Guimarães ML et al. Proteinases as virulence factors in Leishmania spp. infection in mammals. Parasit Vectors, 5, 160 (2012). Carrión J, Folgueira C, Soto M et al. Leishmania infantum HSP70-II null mutant as candidate vaccine against leishmaniasis: a preliminary evaluation. Parasit Vectors, 4(1), 150 (2011). Anand S, Madhubala R. Genetically Engineered Ascorbic acid-deficient Live Mutants of Leishmania donovani induce long lasting Protective Immunity against Visceral Leishmaniasis. Scientific reports, 5 (2015). Ghaffarifar F, Jorjani O, Mirshams M et al. Photodynamic therapy as a new treatment of cutaneous leishmaniasis. Eastern Mediterranean Health Journal, 12 (6), 902-908, 2006. Kumari S, Samant M, Khare P et al. Photodynamic vaccination of hamsters with inducible suicidal mutants of Leishmania amazonensis elicits immunity against visceral leishmaniasis. European journal of immunology, 39(1), 178-191 (2009). Davoudi N, Khamesipour A, Mahboudi F et al. A dual drug sensitive L. major induces protection without lesion in C57BL/6 mice. PLoS neglected tropical diseases, 8(5) (2014).

27.

** 28.

29.

30.

31.

Downloaded by [] at 03:00 01 March 2016

32.

*

33. 34. 35.

36. 37.

* 38.

* 39.

40.

41.

Breton M, Tremblay MJ, Ouellette M et al. Live nonpathogenic parasitic vector as a candidate vaccine against visceral leishmaniasis. Infection and immunity, 73(10), 63726382 (2005). This article is the first one which introduces L. tarentolae as non pathogenic parasite. 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, 28(1), 53-62 (2009). Zahedifard F, Gholami E, Taheri T et al. Enhanced protective efficacy of nonpathogenic recombinant Leishmania tarentolae expressing cysteine proteinases combined with a sand fly salivary antigen. PLoS Negl Trop Dis, 8(3), e2751 (2014). Saljoughian N, Taheri T, Zahedifard F et al. Development of novel prime-boost strategies based on a tri-gene fusion recombinant L. tarentolae vaccine against experimental murine visceral Leishmaniasis. PLoS Negl Trop Dis, 7(4), e2174 (2013). Shahbazi M, Zahedifard F, Taheri T et al. Evaluation of Live Recombinant Nonpathogenic Leishmania tarentolae Expressing Cysteine Proteinase and A2 Genes as a Candidate Vaccine against Experimental Canine Visceral Leishmaniasis. PloS one, 10(7), e0132794 (2015). Katebi A, Gholami E, Taheri T et al. Leishmania tarentolae secreting the sand fly salivary antigen PpSP15 confers protection against Leishmania major infection in a susceptible BALB/c mice model. Molecular immunology, 67(2), 501-511 (2015). It is the first report for using L. tarentulae as live nonpathogenic Leishmania for delivering combination of sandfly salivary protein and Leishmania antigens to improve vaccination strategies against cutaneous leishmaniasis. Schwendener RA. Liposomes as vaccine delivery systems: a review of the recent advances. Therapeutic advances in vaccines, 2(6), 159-182 (2014). Rao M, Alving CR. Delivery of lipids and liposomal proteins to the cytoplasm and Golgi of antigen-presenting cells. Advanced drug delivery reviews, 41(2), 171-188 (2000). Portuondo DLF, Ferreira LS, Urbaczek AC et al. Adjuvants and delivery systems for antifungal vaccines: Current state and future developments. Medical mycology, myu045 (2014). Akbarzadeh A, Rezaei-Sadabady R, Davaran S et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett, 8(1), 102 (2013). Badiee A, Khamesipour A, Samiei A et al. The role of liposome size on the type of immune response induced in BALB/c mice against leishmaniasis: rgp63 as a model antigen. Experimental parasitology, 132(4), 403-409 (2012). This article shows the large size (≥P400 nm) liposomes containing Leishmania antigen can induce protective Th1 immune responses. Afrin F, Rajesh R, Anam K et al. Characterization of Leishmania donovani antigens encapsulated in liposomes that induce protective immunity in BALB/c mice. Infection and immunity, 70(12), 6697-6706 (2002). It is the first report that shows that cationic liposomes louded with Leishmania antigen can induce protective immune responses. Bhowmick S, Ravindran R, Ali N. Leishmanial antigens in liposomes promote protective immunity and provide immunotherapy against visceral leishmaniasis via polarized Th1 response. Vaccine, 25(35), 6544-6556 (2007). Badiee A, Jaafari MR, Khamesipour A et al. The role of liposome charge on immune response generated in BALB/c mice immunized with recombinant major surface glycoprotein of Leishmania (rgp63). Experimental parasitology, 121(4), 362-369 (2009). Bhowmick S, Mazumdar T, Sinha R et al. Comparison of liposome based antigen delivery systems for protection against Leishmania donovani. Journal of Controlled Release, 141(2), 199-207 (2010).

42.

43. 44.

45.

46.

Downloaded by [] at 03:00 01 March 2016

47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

McConville MJ, Bacic A, Mitchell GF et al. Lipophosphoglycan of Leishmania major that vaccinates against cutaneous leishmaniasis contains an alkylglycerophosphoinositol lipid anchor. Proceedings of the National Academy of Sciences, 84(24), 8941-8945 (1987). Gregory G. Liposome technology (CRC press, 2006). Badiee A, Jaafari MR, Khamesipour A et al. Enhancement of immune response and protection in BALB/c mice immunized with liposomal recombinant major surface glycoprotein of Leishmania (rgp63): the role of bilayer composition. Colloids and Surfaces B: Biointerfaces, 74(1), 37-44 (2009). Mazumdar T, Anam K, Ali N. A mixed Th1/Th2 response elicited by a liposomal formulation of Leishmania vaccine instructs Th1 responses and resistance to Leishmania donovani in susceptible BALB/c mice. Vaccine, 22(9), 1162-1171 (2004). Shimizu Y, Yamakami K, Gomi T et al. Protection against Leishmania major infection by oligomannose-coated liposomes. Bioorganic & medicinal chemistry, 11(7), 1191-1195 (2003). Shimizu Y, Takagi H, Nakayama T et al. Intraperitoneal immunization with oligomannose‐coated liposome‐entrapped soluble leishmanial antigen induces antigen‐ specific T‐helper type immune response in BALB/c mice through uptake by peritoneal macrophages. Parasite immunology, 29(5), 229-239 (2007). Chavoshian O, Biari N, Badiee A et al. Sphingomyelin liposomes containing soluble Leishmania major antigens induced strong Th2 immune response in BALB/c mice. Iranian journal of basic medical sciences, 16(9), 965 (2013). Sharma SK, Dube A, Nadeem A et al. Non PC liposome entrapped promastigote antigens elicit parasite specific CD8+ and CD4+ T-cell immune response and protect hamsters against visceral leishmaniasis. Vaccine, 24(11), 1800-1810 (2006). Jaafari MR, Ghafarian A, Farrokh-Gisour A et al. Immune response and protection assay of recombinant major surface glycoprotein of Leishmania (rgp63) reconstituted with liposomes in BALB/c mice. Vaccine, 24(29), 5708-5717 (2006). Jaafari MR, Badiee A, Khamesipour A et al. The role of CpG ODN in enhancement of immune response and protection in BALB/c mice immunized with recombinant major surface glycoprotein of Leishmania (rgp63) encapsulated in cationic liposome. Vaccine, 25(32), 6107-6117 (2007). Russell DG, Alexander J. Effective immunization against cutaneous leishmaniasis with defined membrane antigens reconstituted into liposomes. The Journal of Immunology, 140(4), 1274-1279 (1988). Shargh VH, Jaafari MR, Khamesipour A et al. Cationic liposomes containing soluble Leishmania antigens (SLA) plus CpG ODNs induce protection against murine model of leishmaniasis. Parasitology research, 111(1), 105-114 (2012). Badiee A, Jaafari MR, Khamesipour A. Leishmania major: immune response in BALB/c mice immunized with stress-inducible protein 1 encapsulated in liposomes. Experimental parasitology, 115(2), 127-134 (2007). Badiee A, Jaafari MR, Samiei A et al. Coencapsulation of CpG oligodeoxynucleotides with recombinant Leishmania major stress-inducible protein 1 in liposome enhances immune response and protection against leishmaniasis in immunized BALB/c mice. Clinical and Vaccine Immunology, 15(4), 668-674 (2008). Afrin F, Ali N. Adjuvanticity and protective immunity elicited by Leishmania donovani antigens encapsulated in positively charged liposomes. Infection and immunity, 65(6), 2371-2377 (1997). Mazumdar T, Anam K, Ali N. Influence of phospholipid composition on the adjuvanticity and protective efficacy of liposome-encapsulated Leishmania donovani antigens. Journal of Parasitology, 91(2), 269-274 (2005).

*

58.

59.

60.

61.

Downloaded by [] at 03:00 01 March 2016

62.

63.

64.

65.

66.

67.

68. 69.

70.

71.

72.

This article shows that chemical composition of liposome affects to the type of immune response induction and liposomes contains DSPC induce protective immunity against leishmaniasis. Ravindran R, Bhowmick S, Das A et al. Comparison of BCG, MPL and cationic liposome adjuvant systems in leishmanial antigen vaccine formulations against murine visceral leishmaniasis. BMC microbiology, 10(1), 181 (2010). Afrin F, Anam K, Ali N. Induction of partial protection against Leishmania donovani by promastigote antigens in negatively charged liposomes. Journal of Parasitology, 86(4), 730-735 (2000). Mazumder S, Ravindran R, Banerjee A et al. Non-coding pDNA bearing immunostimulatory sequences co-entrapped with leishmanial antigens in cationic liposomes elicits almost complete protection against experimental visceral leishmaniasis in BALB/c mice. Vaccine, 25(52), 8771-8781 (2007). Bhowmick S, Ravindran R, Ali N. gp63 in stable cationic liposomes confers sustained vaccine immunity to susceptible BALB/c mice infected with Leishmania donovani. Infection and immunity, 76(3), 1003-1015 (2008). Didier E, Bhowmick S, Ali N. Identification of novel Leishmania donovani antigens that help define correlates of vaccine-mediated protection in visceral leishmaniasis. PloS one, 4(6), Article ID e5820, 5810 pages (2009). Bhowmick S, Mazumdar T, Ali N. Vaccination route that induces transforming growth factor β production fails to elicit protective immunity against Leishmania donovani infection. Infection and immunity, 77(4), 1514-1523 (2009). Mazumder S, Maji M, Ali N. Potentiating effects of MPL on DSPC bearing cationic liposomes promote recombinant GP63 vaccine efficacy: high immunogenicity and protection. PLoS Negl Trop Dis, 5(12), e1429 (2011). Ravindran R, Maji M, Ali N. Vaccination with Liposomal Leishmanial Antigens Adjuvanted with Monophosphoryl Lipid–Trehalose Dicorynomycolate (MPL-TDM) Confers Long-Term Protection against Visceral Leishmaniasis through a Human Administrable Route. Molecular pharmaceutics, 9(1), 59-70 (2011). Das A, Ali N. Combining cationic liposomal delivery with MPL-TDM for cysteine protease cocktail vaccination against Leishmania donovani: evidence for antigen synergy and protection. PLoS Negl Trop Dis, 8(8), e3091 (2014). Nagill R, Kaur S. Enhanced efficacy and immunogenicity of 78kDa antigen formulated in various adjuvants against murine visceral leishmaniasis. Vaccine, 28(23), 4002-4012 (2010). Hejazi H, Tasbihi M, Jaafari M et al. The role of liposomal CpG ODN on the course of L. major infection in BALB/C mice. Iranian journal of parasitology, 5(1), 47 (2010). Alavizadeh SH, Badiee A, Khamesipour A et al. The role of liposome–protamine–DNA nanoparticles containing CpG oligodeoxynucleotides in the course of infection induced by Leishmania major in BALB/c mice. Experimental parasitology, 132(3), 313-319 (2012). Sohrabi Y, Jaafari MR, Mohammadi A et al. Evaluation of immune response against leishmaniasis in resistance C57 BL-6 mice immunized with liposomes containing autoclaved Leishmania major with BCG. Cellular and Molecular Biology Letters, (10) (2005). Maheshwari C, Pandey R, Chaurasiya A et al. Non-ionic surfactant vesicles mediated transcutaneous immunization against hepatitis B. International immunopharmacology, 11(10), 1516-1522 (2011). LezamaDávila CM. Vaccination of C57BL/10 mice against cutaneous leishmaniasis. Use of purified gp63 encapsulated into niosomes surfactants vesicles: a novel approach. Memórias do Instituto Oswaldo Cruz, 94(1) (1999).

73.

74.

75. 76.

77.

78.

Downloaded by [] at 03:00 01 March 2016

79. 80.

81.

82.

83.

84.

85.

86.

87.

88.

Pardakhty A, Shakibaie M, Daneshvar H et al. Preparation and evaluation of niosomes containing autoclaved Leishmania major: a preliminary study. Journal of microencapsulation, 29(3), 219-224 (2012). Bolhassani A, Shirbaghaee Z, Agi E et al. VLP production in Leishmania tarentolae: A novel expression system for purification and assembly of HPV16 L1. Protein expression and purification, 116, 7-11 (2015). Gamvrellis A, Leong D, Hanley JC et al. Vaccines that facilitate antigen entry into dendritic cells. Immunology and cell biology, 82(5), 506-516 (2004). Zhou G, Ma Y, Jia P et al. Enhancement of IL-10 bioactivity using an IL-10 peptidebased vaccine exacerbates Leishmania major infection and improves airway inflammation in mice. Vaccine, 28(7), 1838-1846 (2010). Liu X, Siegrist S, Amacker M et al. Enhancement of the immunogenicity of synthetic carbohydrates by conjugation to virosomes: a leishmaniasis vaccine candidate. ACS chemical biology, 1(3), 161-164 (2006). Muller RH, Mader K, Gohla S. Solid lipid nanoparticles (SLN) for controlled drug delivery-a review of the state of the art. European journal of pharmaceutics and biopharmaceutics, 50(1), 161-177 (2000). Mehnert W, Mäder K. Solid lipid nanoparticles: production, characterization and applications. Advanced drug delivery reviews, 47(2), 165-196 (2001). Doroud D, Vatanara A, Zahedifard F et al. Cationic solid lipid nanoparticles loaded by cysteine proteinase genes as a novel anti-leishmaniasis DNA vaccine delivery system: characterization and in vitro evaluations. Journal of Pharmacy and Pharmaceutical Sciences, 13(3), 320-335 (2010). Doroud D, Zahedifard F, Vatanara A et al. Delivery of a cocktail DNA vaccine encoding cysteine proteinases type I, II and III with solid lipid nanoparticles potentiate protective immunity against Leishmania major infection. Journal of Controlled Release, 153(2), 154-162 (2011). Seeballuck F, Lawless E, Ashford MB et al. Stimulation of triglyceride-rich lipoprotein secretion by polysorbate 80: in vitro and in vivo correlation using Caco-2 cells and a cannulated rat intestinal lymphatic model. Pharmaceutical research, 21(12), 2320-2326 (2004). Doroud D, Zahedifard F, Vatanara A et al. Cysteine proteinase type I, encapsulated in solid lipid nanoparticles induces substantial protection against Leishmania major infection in C57BL/6 mice. Parasite immunology, 33(6), 335-348 (2011). Doroud D, Zahedifard F, Vatanara A et al. C-terminal domain deletion enhances the protective activity of cpa/cpb loaded solid lipid nanoparticles against Leishmania major in BALB/c mice. PLoS Negl Trop Dis, 5(7), e1236 (2011). Saljoughian N, Zahedifard F, Doroud D et al. Cationic solid–lipid nanoparticles are as efficient as electroporation in DNA vaccination against visceral leishmaniasis in mice. Parasite immunology, 35(12), 397-408 (2013). Shahbazi M, Zahedifard F, Saljoughian N et al. Immunological comparison of DNA vaccination using two delivery systems against canine leishmaniasis. Veterinary parasitology, 212(3), 130-139 (2015). Tafaghodi M, Tabassi SAS, Jaafari MR. Induction of systemic and mucosal immune responses by intranasal administration of alginate microspheres encapsulated with tetanus toxoid and CpG-ODN. International journal of pharmaceutics, 319(1), 37-43 (2006). Tafaghodi M, Sajadi Tabasi SA, Payan M. Alginate microsphere as a delivery system and adjuvant for autoclaved Leishmania major and Quillaja saponin: preparation and characterization. Iranian Journal of Pharmaceutical Sciences, 3(2), 61-68 (2007).

89.

90. 91.

92.

93.

Downloaded by [] at 03:00 01 March 2016

94.

95.

96.

97.

98.

99.

100. 101.

102.

103.

104. 105.

Tafaghodi M, Eskandari M, Khamesipour A et al. Alginate microspheres encapsulated with autoclaved Leishmania major (ALM) and CpG-ODN induced partial protection and enhanced immune response against murine model of leishmaniasis. Experimental parasitology, 129(2), 107-114 (2011). Lou PJ, Cheng WF, Chung YC et al. PMMA particle‐mediated DNA vaccine for cervical cancer. Journal of biomedical materials research Part A, 88(4), 849-857 (2009). Zarrati S, Maleki F, Mahdavi M et al. Humoral immune responses in DNA vaccine formulated with poly (methyl methacrylate) against Leishmania major. Journal of Entomology and Zoology Studies, 2 (5): 201-206 (2014) . Sharma R, Agrawal U, Mody N et al. Polymer nanotechnology based approaches in mucosal vaccine delivery: Challenges and opportunities. Biotechnology advances, 33(1), 64-79 (2015). Shen H, Ackerman AL, Cody V et al. Enhanced and prolonged cross‐presentation following endosomal escape of exogenous antigens encapsulated in biodegradable nanoparticles. Immunology, 117(1), 78-88 (2006). Tafaghodi M, Khamesipour A, Jaafari M. Immunization against leishmaniasis by PLGA nanospheres loaded with an experimental Autoclaved Leishmania major (ALM) and Quillajasaponins. Tropical Biomedicine, 27(3), 639-650 (2010). Tafaghodi M, Khamesipour A, Jaafari MR. Immunization against leishmaniasis by PLGA nanospheres encapsulated with autoclaved Leishmania major (ALM) and CpG-ODN. Parasitology research, 108(5), 1265-1273 (2011). Santos DM, Carneiro MW, de Moura TR et al. Towards development of novel immunization strategies against leishmaniasis using PLGA nanoparticles loaded with kinetoplastid membrane protein-11. International journal of nanomedicine, 7, 2115 (2012). Costa Lima SA, Resende M, Silvestre R et al. Characterization and evaluation of BNIPDaoct-loaded PLGA nanoparticles for visceral leishmaniasis: in vitro and in vivo studies. Nanomedicine, 7(12), 1839-1849 (2012). Sjölander A, Baldwin TM, Curtis JM et al. Induction of a Th1 immune response and simultaneous lack of activation of a Th2 response are required for generation of immunity to leishmaniasis. The Journal of Immunology, 160(8), 3949-3957 (1998). Sjölander A, Baldwin TM, Curtis JM et al. Vaccination with recombinant parasite surface antigen 2 from Leishmania major induces a Th1 type of immune response but does not protect against infection. Vaccine, 16(20), 2077-2084 (1998). Papadopoulou G, Karagouni E, Dotsika E. ISCOMs vaccine against experimental leishmaniasis. Vaccine, 16(9), 885-892 (1998). Mehravaran A, Jaafari MR, Jalali SA et al. The role of surface charge of ISCOMATRIX nanoparticles on the type of immune response generated against Leishmaniasis in BALB/c mice. Nanomedicine Journal, 2(4), 249-260 (2015). Coler RN, Hudson T, Hughes S et al. Vaccination Produces CD4 T Cells with a Novel CD154–CD40-Dependent Cytolytic Mechanism. The Journal of Immunology, 195(7), 3190-3197 (2015). Firouzmand H, Badiee A, Khamesipour A et al. Induction of protection against leishmaniasis in susceptible BALB/c mice using simple DOTAP cationic nanoliposomes containing soluble Leishmania antigen (SLA). Acta tropica, 128(3), 528-535 (2013). Smith DM, Simon JK, Baker Jr JR. Applications of nanotechnology for immunology. Nature Reviews Immunology, 13(8), 592-605 (2013). Peters NC, Bertholet S, Lawyer PG et al. Evaluation of recombinant Leishmania polyprotein plus GLA-SE vaccines against sand fly-transmitted Leishmania major in C57Bl/6 mice. Journal of immunology (Baltimore, Md.: 1950), 189(10), 4832 (2012).

Downloaded by [] at 03:00 01 March 2016

Fig1: Schematic presentation of different delivery system on Th1 and Th2 response and inducing long life protection

Fig2: Live and chemical delivery system involved in vaccine development against leishmaniasis

Downloaded by [] at 03:00 01 March 2016

Abbreviations: GLA-SE, glucopyranosyl lipid adjuvant-stable emulsion; PMMA, Poly methyl methacrylate; PLGA, poly(lactic-co-glycolic acid); ISCOMS, Immune Stimulating Complexes; cSLN, Cationic Solid Lipid Nanoparticles.

Challenge parasite

Live vaccine

Antigen

Experimental model

1

L. donovani

Vaccinia virus

KMP11

Hamster/BALB/c

2

L. major

Influenza virus

LACK158–173

BALB/c

L. major

Canine distemper virus (CDV)

3

LACK/TSA/LMSTI1

Dog

Table1: Live Viral delivery systems in vaccination against Leishmania parasite

Reference [2] Guha et al.(2013) [4] Kedzierska et al. (2012) [5] Miura et al. (2015)

Challenge parasite

Antigen

Experimental model

1

L. major

Lactococcus lactis

LACK/IL-12 knock in

BALB/c

2

L. donovani

Lactococcus lactis

A2 Knock in

BALB/c

3

L. major

Listeria monoctogenes

LJM11

C57BL/6

4

L. major

Salmonella typhi

LinJ08.1190/LinJ23.0410

BALB/c

BCG

-

BALB/c

5 Downloaded by [] at 03:00 01 March 2016

Live vaccine

L. tropica

6

L. major /L. mexicana

BCG

Gp63

BALB/c CBA

7

L. major

BCG

Gp63

BALB/c

8

L. chagasi

BCG

LCR1 knock in

BALB/c

Table 2: Live bacterial delivery systems in vaccination against Leishmania infection

Reference [6,7] Hugentobler et al. (2012) [8] Yam K et al. (2011) [9] Abdallah et al. (2014) [11] Schroeber et al. (2011) [12] Weintraub. J. et.al. [13] Connell ND. et al. [14] Abdelhak. et al. [15] Streit, J. A., et al.

Downloaded by [] at 03:00 01 March 2016

Challenge parasite

Live vaccine

gene

Experimental model

Kind of vehicle

Reference [18,28] Dey et al. (2014) [19] Fiuza J et al. (2015) [22] Carrion et al. (2011)

1

L. mexicana

L. donovani

LdCen-/-/ Ldp27 -/-

BALB/c

Attenuated parasite

2

L. infantum

L. donovani

Centrin

Dog

Attenuated parasite

3

L. major

L. infantum

HSP70 II

BALB/c

Null Mutant parasite

4

L.donovani

L.donovani

ALO

BALB/c

Attenuated parasite

[23] Anand et al. (2015)

5

L. donovani

L. amazoniensis

delta-aminolevulinate (ALA) dehydratase and porphobilinogendeaminase

Hamster

Recombinant parasite

[18,25] Kumari et al. (2009)

C57BL/6

Recombinant parasite

[26,30] Davoudi et al. (2014) [26,27] Breton et al. (2005)

6

L. major

7

L. donovani

L. tarentolae

Whole parasite

BALB/c

Recombinant parasite

8

L. infantum

L. tarentolae

A2 Knock in

BALB/c

Recombinant parasite

[28,31] Mizbani et al. (2009)

BALB/c

Recombinant parasite

[25,29] Zahedifard et al. (2014) [27,30] Saljoughian et al. (2013) [31] Shahbazi et al. (2015)

9

L. major

L. major

L. tarentolae

Lmtkcd+/+

CPA/CPB/PpSP15

10

L. infantum

L. tarentolae

A2-CPA-CPB -CTE

BALB/c

Recombinant parasite

11

L. infantum

L. tarentolae

A2-CPA-CPB -CTE

Dog

Recombinant parasite

12

L. major

L. tarentolae

PpSP15

BALB/c

Recombinant parasite

Downloaded by [] at 03:00 01 March 2016

Table3: Live Leishmania vaccine as delivery system against leishmaniasis

[23,32] Katebi et al. (2015)

Type of chemical delivery system: Composition Liposome: egg lecithin/cholesterol, 7:2

Downloaded by [] at 03:00 01 March 2016

1

2

3

4

5

6

egg lecithin/cholesterol/stearylamine, 7:2:2 Liposome: egg lecithin/cholesterol/stearylamine, 7:2:2 Liposome: egg lecithin/cholesterol/stearylamine, 7:2:2 Liposome: egg lecithin/cholesterol/stearylamine, 7:2;2 Liposome: egg lecithin/cholesterol/stearylamine, 7;2:2 Liposome: lecithin/cholesterol/stearylamine, 7:1:2 (DRV)

IS

Target Antigen

Route of Injection

Challenge with

Experiment al model

Outcome comment

Reference

-

LAg

i.p.

L. donovani

BALB/c

Cationic liposome is protective.

[38] Afrin et al. (2002)

[63] Bhowmick et al. (2009)

L. donovani

BALB/c

Mice immunized i.p. and i.v. with LAg, either free or encapsulated in liposomes, was protected against challenge.

-

LAg

i.p., i.v., s.c., and i.m.

-

LAg

i.p.

L. donovani

Hamster / BALB/c

protection

[56] Afrin and Ali (1997)

pDNA

SLA

i.p.

L. donovani

BALB/c

protection

[60] Mazumder et al. (2007)

Comparison of BCG, MPL, and TDM Comparison of MPL-A, liposomal encapsulatio n, rIL-12, ALD and

LAg

i.p.

L. donovani

BALB/c

protection

[58] Ravindran et al. (2010)

naive 78 kDa antigen of L. donovani

s.c.

L. major

BALB/c

protection

[67] Nagill (2010)

Freund’s adjuvant Liposome: egg lecithin/cholesterol, 7:2

Downloaded by [] at 03:00 01 March 2016

7

positively egg lecithin/cholesterol/stearylamine, 7:2:2

-

SLA

i.p.

L. donovani

BALB/c

protection

[39] Bhowmick et al. (2007)

protection

[50] Jaafari et al (2006)

protection

[49] Sharma et al (2006)

negative egg lecithin/cholesterol/phosphatidic acid, 7:2:2 8

9

Liposome: egg lecithin/cholesterol, 1:1 Liposome: egg lecithin (PC)/ cholesterol, 7:3

-

-

rGP63

sLAg

s.c.

i.p.

L. major

BALB/c

L. donovani

hamster / BALB/c

non-phosphatidylcholine (nonPC) liposomes (escheriosomes) 10

Liposome: DSPC/cholesterol, 7:2

-

LAg

i.p.

L. donovani

BALB/c

moderate protection

11

Liposome: DPPC/cholesterol, 2:1

CpG ODNs

rLmSTI1

s.c.

L. major

BALB/c

complete protection

12

Liposome: DPPC/cholesterol, 2:1 (DRV) with different sizes (100, 400, 1000 nm)

-

rGP63

s.c.

L. major

BALB/c

The large size (≥P400 nm) liposomes induce protective Th1

[45] Mazumdar et al (2004) [55] Badiee et al (2008) [37] Badiee et al (2012)

immune responses. Liposome: DSPC/cholesterol, 7:2 13

DPPC/cholesterol, 7:2

-

LAg

i.p.

L. donovani

hamster

Liposomes contains DSPC induce protective immunity.

[57] Mazumdar et al (2005)

Downloaded by [] at 03:00 01 March 2016

DMPC/cholesterol, 7:2 14

Liposome: DSPC/cholesterol, 2:1

-

rLmSTI1

s.c.

L. major

BALB/c

protection

15

Liposome: DSPC/cholesterol/stearylamine, 7:2:2

-

Naïve L. donovani GP63

i.p.

L. donovani

BALB/c

protection

16

Liposome: DSPC/cholesterol/stearylamine, 7:2:2

-

L.donovani: polypeptides LD91, LD72, LD51, LD31

i.p.

L. donovani

BALB/c

protection

MPL-TDM

L. donovani rCPs (a, b, c)

s.c.

L. donovani

hamster

complete protection

MPL-TDM

rGP63

s.c.

L. donovani

BALB/c

protection

BALB/c

Liposomes that made with MLV and DRV methods can induce protective immunity.

BALB/c

Liposomes containing DSPC and DPPC can induce protective immunity.

17

18

19

Liposome: DSPC/cholesterol/stearylamine, 7:2:2 Liposome: DSPC/cholesterol/stearylamine, 7:2:2 Liposome: DSPC/cholesterol/stearylamine, 7:2:2

-

LAg

i.p.

L. donovani

(MLV, DRV, and REV)

20

Liposome: DSPC/cholesterol, 2:1 Tc 54 ◦C DPPC/cholesterol, 2:1 Tc 41 ◦C

-

rGP63

s.c.

L. major

[54] Badiee et al (2007) [61] Bhowmick et al (2008) [62] Bhowmick and Ali (2009) [66] Das and Ali (2014) [64] Mazumder et al (2011) [41] Bhowmick et al (2010)

[44] Badiee et al (2009)

EPC/cholesterol, 2:1 Tc 0 ◦C Liposome: Neutral :DPPC/cholesterol, 2:1

Downloaded by [] at 03:00 01 March 2016

21

Positively: DPPC/cholesterol/DDAB, 2:1:1

-

rGP63

s.c.

L. major

BALB/c

Neutral liposome induced protective Th1 responses

[40] Badiee et al (2009)

CpG ODNs

rGP63

s.c.

L. major

BALB/c

protection

[51] Jaafari et al (2007)

-

SLA

i.p.

L. major

BALB/c

protection

[46] Shimizu et al (2003)

-

SLA

i.p.

L. major

BALB/c

protection

[47] Shimizu et al (2007)

-

SLA

s.c.

L. major

BALB/c

none protection

CpG ODNs

SLA

s.c.

L. major

BALB/c

complete protection

CpG ODNs

live L. major

s.c.

L. major

BALB/c

partial protection

-

SLA

s.c.

L. major

BALB/c

protection

Negatively: DPPC/cholesterol/DCP, 2:1:1 (DRV) 22

23

Liposome: DPPC/cholesterol/DDAB, 2:1:1 Liposome: DPPC/cholesterol/DPPE DPPC/cholesterol/Man5-DPPE Liposome: DPPE/cholesterol/DPPC

24

25

26

27 28

DPPC/cholesterol/Man5-DPPE DPPC/cholesterol/Man3-DPPE Liposome: Sphingomyelin/cholesterol, 13:1 (detergent removal method) Liposome: DOTAP/cholesterol, 1:1 (lipid film method) Liposome: DOTAP/cholesterol, 1:1 (lipid film method) Liposome:

[48] Chavoshian et al (2013) [53] Shargh et al (2012) [69] Alavizadeh et al (2012) [103]

29

Downloaded by [] at 03:00 01 March 2016

30

31

32

33

DOTAP/cholesterol, 1:1 (MLV) cSLN: (cetyl palmitate /cholesterol/DOTAP/Tween 80, 3.2:1) melt emulsification method cSLN: (cetyl palmitate /cholesterol/DOTAP/Tween 80, 3.2:1) melt emulsification method cSLN: (cetyl palmitate /cholesterol/DOTAP/Tween 80, 3.2:1) melt emulsification method cSLN: (cetyl palmitate /cholesterol/DOTAP/Tween 80, 3.2:1) melt emulsification method cSLN: (cetyl palmitate/cholesterol/DOTAP/T ween 80, 3.2:1) melt emulsification method

Firouzmand et al. (2013)

-

pDNA of L. major cpa/b/c

s.c.

L. major

BALB/c

protection

[81] Doroud et al. (2011)

-

pDNA of L. major cpa/b CTE

i.p.

L. major

C57BL⁄6

protection

[83] Doroud et al. (2011)

-

pDNA of L. major cpa/b CTE

s.c.

L. major

BALB/c

protection

[84] Doroud et al. (2011)

-

pDNA of a2cpa/cpb_CTE

s.c.

L. infantum

BALB/c

partial protection

[85] Salgoughian et al. (2013)

-

pDNA of a2cpa/cpb_CTE

s.c.

L. infantum

Dog

protection

[86] Shahbazi et al. (2015) [72]

34

Niosome: (single-alkyl chain, non-ionic surfactant/cholesterol)

-

Naïve L. mexicana GP63

s.c.

L. mexicana

C57BL/10

Partial protection

35

Niosome: (sorbitan esters/cholesterol/cetyl trimethyl ammonium bromide)

-

ALM

s.c.

L. major

BALB/c

Moderate protection

LezamaDávi la CM (1999) [73] Abbas

Pardakhty (2012)

Downloaded by [] at 03:00 01 March 2016

36

influenza virosomes (IRIVs): (egg PC/PE/ glycan-PE conjugate/HA of inactivated influenza A/Singapore virus, 16:4:1:2)

-

Leishmania carbohydrates (LPG)

i.m.

-

BALB/c

Elicit T-cell dependent antibody responses Against oligosaccharide antigens

[77] Liu et al. (2006) [89] Tafaghodi et al.(2011) [91] Zarrati et al. (2014) [94] Tafaghodi et al. (2010) [95] Tafaghodi et al. (2011)

37

Alginate microsphere

CpG ODNs

ALM

s.c.

L. major

BALB/c

protection

38

PMMA

-

pDNA: tsa

i.m.

L. major

BALB/c

Elicit humoral immune responses

39

PLGA nanospheres

-

ALM

s.c.

L. major

BALB/c

protection

40

PLGA nanospheres

CpG ODNs

ALM

s.c.

L. major

BALB/c

protection

i.d.

L. braziliensis + L. intermedia SGS

BALB/c

protection

[96] Santos et al. (2012)

i.p.

L. major

C3H/He

None protection

[98,99] Sjo¨lander et al.(1998)

41

PLGA nanoparticles

-

pDNA or recombinant protein of L. infantum chagasi KMP11

42

ISCOMs: (phosphatidylcholine/ISCOPRE P 703 [Quil A]/cholesterol, 1:3:1)

-

pDNA of psa-2

43

ISCOMs: (triterpcnoids/cholesterol/phosph atidylcholine/MEGA10/QuilA)

-

Naïve L. major GP63

i.p.

L. major

BALB/c

partial protection

44

ISCOMs:

-

SLA

s.c.

L. major

BALB/c

None protection

[100] Papadopoulo u et al.(1998) [101]

(DSPC or DOTAP/ Quil A/cholesterol, 2:2:1) 45

Downloaded by [] at 03:00 01 March 2016

46

47

48

49

GLA-SE

GLA-SE

GLA-SE

GLA-SE

GLA-SE

GLA

LEISH-F3 polyprotein

s.c. at the base of the tail

L. donovani and L. infantum

C57BL/6 and BALB/c

Protection immune responses

GLA

LEISH-F3 polyprotein

-

-

Human clinical trial phase I

safety and efficacy of vaccine was confirmed in human

GLA

GLA

GLA

KSAC and L110F

SODB1, Pxn4

KSAC

Table4: chemical delivery system in vaccine development against leishmaniasis

s.c.

s.c.

s.c.

L. major

L. donovani

L. major

BALB/c

L110F-GLA induces partial protection while KSAC-GLA induces strong protection

Mehravaran et al. (2015) [1] Coler RN et al.(2015) [1] Coler RN et al.(2015)

[102] R Gomes et al. (2012)

BALB/c

Protection immune responses

[103,104] NS Daifalla et al.(2011, 2012)

C57BL/6

strong protection against needle challenge

[105] Peters et al. (2012)

Table Abbreviation:

Downloaded by [] at 03:00 01 March 2016

ALD: Autoclaved Leishmania donovani antigen; ALM: autoclaved L. major; ALO: Arabino-1, 4-lactone oxidase; DOTAP : 1,2-dioleoyl-3-trimethylammonium-propane; gp63: glycoprotein 63; GLA-SE: Glucopyranosyl lipid A in stable oil-in-water nanoemulsion; HSP: heat shock protein; IS: Immunostimulant; i.m.: intramuscularly; i.d.: intradermal; i.p.: intraperitoneally; KMP-11: kinetoplastid membrane protein-11; LACK: Leishmania homologue of activated C kinase; LAg: L. donovani promastigote membrane antigens; LCR1: antigen cloned from amastigote L. chagasi library; LPG: Lipophosphoglycan; MPL-TDM: Monophosphoryl lipid A-trehalose dicorynomycolate; MPL-A: Monophosphoryl lipid A; pDNA: plasmid DNA; pDNA of A2-CPA-CPB_CTE: DNA vaccine harbouring the L. donovani A2 antigen along with L. infantum cysteine proteinases (CPA and CPB without its unusual C-terminal extension); Pxn4: Peroxidoxin4; PLGA: poly(lactic-co-glycolic acid); PpSP15: Phlebotomus papatasi salivary protein15; PSA-2: L. major parasite surface Ag-2; rGP63: recombinant major surface glycoprotein of Leishmania; rLmSTI1: Recombinant L. major stress-inducible protein 1; s.c.: subcutaneous; SGS: salivary gland sonicate; sLAg: L. donovani promastigote soluble antigens; SLA: Soluble Leishmania antigens; SODB1: iron superoxide dismutase B1; TSA: Thiol-Specific Antioxidant Protein;

Delivery systems for Leishmania vaccine development.

Leishmaniasis is a neglected disease and is endemic in tropical and sub-tropical areas worldwide. Lifelong immunity after recovery indicates that vacc...
900KB Sizes 0 Downloads 16 Views