vaccines for leishmaniasis in the fore coming 25 years

16

Click here to load reader

Upload: clarisa-b-palatnik-de-sousa

Post on 30-Oct-2016

230 views

Category:

Documents


5 download

TRANSCRIPT

  • Vaccine (2008) 26, 17091724

    avai lab le at www.sc iencedi rec t .com

    journa l homepage: www.e lsev ier .com/ locate /vacc ine

    REVIEW

    Vacccomi

    Clarisa B. Palatnik-de-Sousa

    Instituto de Microbiologia Prof. Paulo de Goes, Centro de Ciencias da Saude, Universidade Federal do Rio de Janeiro, PO Box68040 CEP 2

    Received 3Available on

    KEYWOHuman vMice andvaccinesLeishmaFirst-, sethird-gePhase I,Adjuvan

    Contents

    IntroduImmunFirst-g

    CorrespoUniversitaria

    E-mail a

    0264-410X/$doi:10.10161941-590 Rio de Janeiro, Brazil

    November 2007; received in revised form 28 December 2007; accepted 11 January 2008line 30 January 2008

    RDSaccines;dog

    ;niasis;cond- andneration;II, III trials;t

    Summary Human vaccination against leishmaniasis using live Leishmania was used in MiddleEast and Russia (19411980). First-generation vaccines, composed by killed parasites inducelow efcacies (54%) and were tested in humans and dogs Phase III trials in Asia and SouthAmerica since 1940. Second-generation vaccines using live genetically modied parasites, orbacteria or viruses containing Leishmania genes, recombinant or native fractions are knownsince the 1990s. Due to the loss of PAMPs, the use of adjuvants increased vaccine efcacies ofthe puried antigens to 82%, in Phase III dog trials. Recombinant second-generation vaccinesand third-generation DNA vaccines showed average values of parasite load reduction of 68% and59% in laboratory animal models, respectively, but their success in eld trials had not yet beenreported. This review is focused on vaccine candidates that show any efcacy against leishma-niasis and that are already in different phase trials. A lot of interest though was generated inrecent years, by the studies going on in experimental models. The promising candidates maynd a place in the forth coming years. Among them most probably are the multiple-gene DNAvaccines that are stable and do not require cold-chain transportation. In the mean time, second-generation vaccines with native antigens and effective adjuvants are likely to be licensed andused in Public Health control programs in the fore coming 25 years. To date, only three vaccineshave been licensed for use: one live vaccine for humans in Uzbekistan, one killed vaccine forhuman immunotherapy in Brazil and a second-generation vaccine for dog prophylaxis in Brazil. 2008 Published by Elsevier Ltd.

    ction ............................................................................................................ 1710ology of leishmaniasis ............................................................................................ 1710eneration vaccines that arrived to clinical assays................................................................. 1712

    ndence address: Instituto de Microbiologia, CCS, UFRJ, Avda Carlos Chagas 373, Caixa Postal 68040, 21941-590 Cidade, Ilha do Fundao, Rio de Janeiro, Brazil. Tel.: +55 21 25626742; fax: +55 21 2560 8344x145.

    ddress: [email protected].

    see front matter 2008 Published by Elsevier Ltd./j.vaccine.2008.01.023ines for leishmaniasis in the foreng 25 years

  • 1710 C.B. Palatnik-de-Sousa

    Candidates for second-generation vaccines ............................................................................. 1712Live vaccines....................................................................................................... 1712Vaccines using recombinant viruses and bacteria as delivery vehicles .............................................. 1713

    Vaccines based on puried Leishmania antigens......................................................................... 1713Recombinant antigens................................................................................................... 1714Candid .....Vaccine .....Synthe .....Conclu .....Ackn .....Refer .....

    Introduc

    Leishmaniaby the bitin 88 counthreatenincaused humneous lesioleishmanianeous leisdisease wivoirs. ThepopulationOn the otantroponootransmitteAmerica [1vaccines. Tand dogs isof chemothimmunocomdemiologicdogs [2]. VLAsia), Leishtum (Meditand L. braL. tropicaleishmaniaLeishmanialent vaccinfor prophyl

    The rsby ProfessoIsrael, whotheir childrintuitivelygle rst lesin future [ulate uninflesions, inhidden [4]asites wasIsrael andgenerationcrude extrdue to uncHIV and th

    paraum credialses ced lee beishmecomn geande offoreuk].ordisignts toe [1e-indAlthccinheyallent thas d

    ts ofccessdedIV trparts, gils arringe tre

    nolates for third-generation vaccines .............................s based on sand y salivary antigens ..........................tic vaccines ....................................................sions and perspectives .........................................owledgements..................................................ences..........................................................

    tion

    sis is caused by parasitic protozoa transmittede of female sand y and is currently endemictries, affecting 12 million people worldwide andg 350 million more. Several species of Leishmaniaan diseases that range form self-healing cuta-ns to fatal visceral leishmaniasis (VL), mucosalsis and diffuse cutaneous leishmaniasis. Cuta-hmaniasis (CL) is an antroponotic or zoonoticth wild rodent, canids and marsupials as reser-difculties in control of the wild reservoir

    led to the development of human vaccines only.her hand, VL, the most severe disease, is anse in India and Central Africa, and a zoonosisd by domestic dogs in the Mediterranean and]; leading to the search of both, human and doghe need for safe prophylactic vaccines for humanmade greater by the drug resistance and toxicityerapy, the increase of the disease incidence inpromised subjects, and the difculties of epi-

    al control based upon sacrice of seropositiveis caused by Leishmania donovani (Africa, India,mania chagasi (America) and Leishmania infan-erranean basin). CL is produced by L. mexicanaziliensis complexes in Americas and L. major,and L. aethiopica in the Old World [1,3]. As allsis are caused by closely phylogenetically relateds species, the development of a unique polyva-e is expected and will be very valuable not onlyaxis but for treatment as well.t vaccine against leishmaniasis was developedr Adler at the Hebrew University of Jerusalem,had observed that mothers of Lebanon exposedens arms to the bite of sand ies because theyknew that the development of a self-healing sin-ion would protect them from the severe disease4]. Therefore, the ancient practice was to inoc-

    sons,inoculregistecacy trvaccinreplacfar haved Leman, rantige[79]The uscodingtaining[1316

    Accare desubjecvaccinvaccinlenge.the vatest, tcial chagainsformedamounhas suforwarPhaseals) asreasoncontrocompavaccin

    Immuected individuals with infectious material fromregions of the body where the scar would be

    . After a method for axenic culture of the par-established, leishmanization became usual inRussia and further evoluted to the use of rst-vaccines composed of whole killed parasites oracts. Leishmanization process was discontinuedontrolled long-lasting skin lesions, the spread ofe use of immunosuppressive drugs, ethical rea-

    Studies oadvancedcell-mediainfection.experimendogs or hum

    Acquirecells [18].CD8+ are m............................................. 1717

    ............................................. 1718

    ............................................. 1718

    ............................................. 1718............................................. 1719............................................. 1719

    site persistence, and difculties in the qualityontrol. Its use at present is limited to one vaccinein Uzbekistan and to live challenge in vaccine ef-in humans in Iran [5]. First-generation Leishmaniaomposed of killed parasites [6] have graduallyishmanization. Second-generation vaccines thusen based on the following: live, genetically modi-ania spp. designed to cause abortive infection inbinant bacteria or viruses carrying Leishmania

    nes, dened synthetic or recombinant subunitsnative fractions puried from parasites [1012].third-generation vaccines [6] that include genesa protective antigen, cloned into a vector con-aryotic promoter, is the more recent approach

    ng to the WHO recommendations, Phase I trialsed to compare vaccinated to placebo treatedassess the immunogenicity and safety of the

    7]. Phase IIa trials are designed to check theuced protection against an experimental chal-ough they are needed in order to standardizee dosage, route and schedule prior to the eldare limited by the questionable value of arti-ge. Phase III trials analyse the vaccine efcacy

    e natural infection. All these assays are per-ouble-blind randomized control trials with smallindividuals (dozens to thousand). If the vaccinefully passed these steps, the formulation can beto registration and industrialization and used inials larger populations (10,000100,000 individu-of national immunization campaigns. For ethical

    ven the benet of the vaccine, no placebo treatede used and the vaccine efcacy is measured bythe incidence of the disease before and after theatment [17].

    ogy of leishmaniasisf anti-Leishmania vaccine candidates havein recent years due to the understanding ofted immunological mechanisms for controllingMost current knowledge, however, is based ontal mouse models and cannot be extrapolated toans.

    d resistance to leishmaniasis is mediated by TCD4+ lymphocytes are crucial for resistance andore involved in memory than as effector cells.

  • Vaccines for leishmaniasis in the fore coming 25 years 1711

    CD4+ CD25+ regulatory T cells are involved in persistence ofL. major infection [19].

    In humans, a correlation between TH1 cell responsesand resistance and healing of cutaneous leishmaniasis wasdescribed with a predominance of cells producing IFN-,while a mixterized muOther studleishmaniapredominale in patieTH1 responpatients [2ease was ddirect correase by LeTNF- wasmixed cytoease by L. cnave andskin test tlevels [27]lead to misis illustratehallmark ono skin tes

    Innateral killer88 [29] hainfection ainnate immrecently duninfectedtest.

    In the mand antigecell respondogs, andmediated sden and aantigen preclass II maImmunosup expressiCD4+ CD25B-7 molecthe CTLA-4CTLA-4 leaexpressionunresponsimouse modsecretion oasites insidthe role ofT CD4+ cel. In additin liver anSoluble facinvolved inis delipidat

    In miceincreases d

    because they fail to localize the periarteriolar lymphoidsheath (PALS) [38]. CCL21 and CCL19 expression by gp38+PALS stromal cells is decreased during infection. For thesereasons DC do not migrate properly in spleen nor do theydestroy the amastigotes. The major mechanism underlying

    iveted imustu

    d TH/6, Cted b- ah th, whst, tse th. In

    ].ananiad bealitor proba2 cyvelsLeishientbclastedof Ig]. A santlineveryE, atheed aG1an

    dogsniner Echtingof t72]redbe date,thantudiatedaccigatiouse

    ntiatheb cyhat hcombum led TH1/TH2 response with IL-4 and IL-10 charac-cocutaneous and chronic cutaneous lesions [20].ies show this mixed cytokine prole in cutaneoussis and in natural resistance, with IL-4 and IL-10ting in early infection and a Type-1 immune pro-nts with older lesions [21], or a main exacerbatedse with high levels of IFN- and TNF- in mucosal2]. In VL, no association of IL-4 with active dis-escribed; however sustained levels of IFN- and aelation between increase in IL-10 and active dis-ishmania donovani were detected [23,24] whilesignicantly elevated in cases of PDKL [25]. Thekine pattern has been conrmed in active dis-hagasi [26], though this study used non-adequateasymptomatic controls selected by a Montenegrohat could itself determine an increase in IFN-or, together with the immunouorescent assay,diagnosis of CL. The problem with this approachd by the identication of higher levels of IL-4, thef CL [28], in control individuals [26]. Furthermore,t was performed on the patient group [26].immunity, toll like receptors, as well as natu-cells, IL-1 and myeloid differentiation factorve been identied in early resistance to thend acquired immunity [30]. The contribution ofunity in human visceral leishmaniasis was also

    escribed [31]. However, this study also selectedand asymptomatic controls by Montenegro skin

    ouse model for VL, Leishmania specic TH2 cellsn presenting cells are involved in suppression of Tses [32]. The DTH response is suppressed in mice,humans and is recovered after cure. Macrophageuppression leads to the increase in parasite bur-ntigen energy, and is linked to either defectivesentation or inhibition of expression of class I andjor histocompatibility complex molecules [28].pression in mice is related to enhanced TGF-

    on [33], IL-10, and the possible participation of+ regulatory cells. There are two receptors forules: the CD28 for T cell activation (TH1) andfor termination of T cell activation. Blockade ofds to resistance to infection, suggesting that theof CTLA-4 plays an important role in maintainingveness in CD4+ T cells during chronical VL in theel [34]. The expression of CTLA-4 results in thef TGF-, which promotes the growing of the par-e the macrophage. In tegumentar leishmaniasis,CTLA-4 is paradoxal, dual [35]. In VL, apoptosis ofls is accompanied by a decrease in IL-2 and IFN-ion, apoptosis is detected in inammatory cellsd spleen during infection with L. donovani [36].tors in serum, such as triglycerides, may also beimmunosuppression. These disappeared if serumed [37].infected with L. donovani, the number of DCuring infection but their distribution is altered

    defectmedia

    Thefor theTH1 an(C57Blmedia12. IFNthroug(iNOS)contrarespondisease[42,43

    HumLeishmtion anabnormolutionIFN- pthe THhigh leof theVL patIgG suassociavation[48,49signic

    CanshowsIgA, Igers ofobservwith Igdiseasenatedthe ca[67] oconicnance[53,69ufactucouldconjugtitresmost svaccinnant vinvestidiseasedifferetor ofand CPshow tthe reinfantlocalization of DCs is TNF- dependent, IL-10-nhibition of CCR7 expression [38].rine model for CL is considered one of the bestdy of the mechanisms controlling the T helper2 cell balance [3941]. In the resistant strains3H, CBA) resolution of the limited infection isy TH1 cells secreting IFN- in response to IL-ctivates the parasiticide activity of macrophagese synthesis of inducible nitric oxide synthaseich leads to production of nitrogen radicals. Inhe susceptible Balb/c mice develop a main TH2at results in progression of lesions and systemicdraining lymph nodes, CD4+ cells secrete IL-4

    kala-azar is characterized by high titers of-specic antibodies appearing soon after infec-fore the development of cellular immunologicalies. The role of these antibodies in disease res-rotection is largely unknown. The TH1 cytokinebly upregulates IgG1 and IgG3 in humans, whiletokines IL-4 and IL-5 stimulate the production ofof IgM, IgE, and IgG isotypes such as IgG4. Analysismania specic Ig isotypes in and IgG subclasses insera revealed elevated levels of IgG, IgM, IgE andses during disease [4447]. Drug resistance waswith a reduction in IgG2 and IgG3. A marked ele-G1, however, was observed in all these patientsuccessful cure corresponded with a decline, mosty, in the levels of IgE, IgG4, and IgG1 [45,46,48].visceral leishmaniasis (CVL), on the other hand,similar characteristics to the human disease [50].nd IgM responses have been shown to be mark-disease [5153]. IgG antibody increase is alsond correlated with symptomatology [50,54,55]subtype associated to susceptibility and severed IgG2 increased in natural resistant or vacci-[15,5666]. IgG2 subtype also predominates inresponse to vaccination against Lyme diseaseinococcus granulosus infection [68]. However,results have been reported, with the predomi-he IgG2 subtype observed in symptomatic dogs. Since all investigations used the same man-anti-dog conjugates, the discrepancy in resultsue to difference in titration mainly of the IgG2which systematically shows signicantly higherthe IgG1 sera [59]. On the other hand, while

    es defending the IgG2 predominance deal withdogs and used puried [59,63,66] or recombi-

    ne antigens for diagnosis [15,57,62,64,65], thens that associate IgG2 to symptomatology andd promastigote crude antigens [15,6971]. Thel afnity of the antigen could be another fac-discrepancy. In dogs vaccinated with the CPastein proteinases, the work of Rafati et al. [15]igher IgG2 than IgG1 titers are detected againstinant antigens but not against the Leishmania

    ysate. While the recombinant antigen interacts

  • 1712 C.B. Palatnik-de-Sousa

    with a dened fraction of sera antibodies, the total lysateinteracts with the whole plethora of antibodies directedagainst the total parasite, masking or diluting the responseagainst the recombinant protein, which is not a majorLeishmania antigen. Another factor of discrepancy couldbe the useand anti-Igexperiencedifcultiesantibodies[72]. Unforavailable.

    When ceasymptomaand TNF-marily TH1asymptomaamastigoteCD8+ incremolecules[75]. In coresponse, wfollowing adecrease inLeishmaniathe co-stimexpressionmal more sand IL-4 haasymptomaease is relagene NRAMof Leishmato the infeMHCII allelinfection wciated to s

    First-genclinical a

    The humanvaccines,started inin Table 1.with no prwere develdirected ag

    Since thtively lowlarge. In Aautoclavedspecies [86claved L. mkilled paraform of stetries thatwhere a coof de Luca eprotozoa pof the para

    ertheless, the LPG complex resists autoclaving [94] and hasbeen implicated in immunogenicity and immunosuppression[95] in the mice model.

    While some protocols have used no adjuvant [86,87,96](Table 1), most prophylactic vaccines used BCG for human

    ] anmo

    es isate6,89ed a91]t incadorly d1). Oof Vity arsvisc

    7] buse I tle [ed athest humTablnd inthatt inso re0]. Ben re

    idat

    acc

    ategknosuc[101orter

    tove inntro insingpes Ieamthorleconpartparaive tnt tochale [luabof different batches of the polyclonal anti-IgG2G1 antisera, which show low repeatibility (ourand Dr MJ Day personal communication). Thesestimulated Dr MJ Day to obtain dog monoclonalthat recognize four different IgG subfractionstunately, these antibodies are not commercially

    llular immune response is present, dogs appeartic and IDR positive, with higher levels of IL-2and a mixed TH1/TH2 response involving pri-mediated by IL-12, IL-18 and IFN-Y [73]. In

    tic dogs, macrophages are capable of killings through the nitric oxide route [74], and aase in peripheral lymphocytes, MHC class IIand receptors CD45RA e CD45RB are also foundntrast, symptomatic dogs show a failed cellularith no IDR or lymphocyte proliferation and the

    dditional characteristics: decrease in monocytes;CD8+ [75] CD4+ and CD21+ B lymphocytes, either-specic or not; antibody increase; deciency ofulatory response; decrease of IFN-; and reducedof MHC class II molecules, which renders the ani-usceptible [66,7580]. The involvement of IL-10s been shown in both symptomatic [81,82] andtic dogs [73]. The susceptibility of the canine dis-ted to a genetic polymorphism. Mutations on theP1, which controls the intralisossomal replicationnia, determine the susceptibility or resistancection [83]. Moreover, a relationship between thees DLA (DRB1, DQA1, DQB1) and the course ofas described with the genotype DLA-DRB1 asso-evere disease [84].

    eration vaccines that arrived tossays

    efcacy trials involving the so called killed-composed of crude total parasite antigens,Brazil in the 1940s [85] and are summarizedThese Phase III efcacy trials were performedevious Phase I or Phase IIa assays. Most trialsoped against CL [8690] while only one [91] wasainst VL.e average vaccine efcacy value (VE) was rela-(54.38%), the number of individuals tested wasmerica, most vaccines used the L. amazonensislysate (ALA) [87,88] or a mixture of autochtonous], while all studies in the Old World used the auto-ajor antigen (ALM) (Table 1). Autoclaving of thesite vaccine was introduced [8992] as the bestrilization and preservation of vaccines in coun-have a rudimentary Biotechnology industry andld-chain for distribution is not feasible. The workt al. [93] proved however, that, as expected for a

    arasite, autoclaving destroys most of the proteinssite and the vaccine loose immunogenicity. Nev-

    [8891The

    vaccincandidity [8obtain[86,89did noin Ecuprobab(Table43.3%mortal

    ThecanineIran [9of Phaavailabobtainmore,agains[100] (vant aagreeapy buorder t[96,10has be[96].

    Cand

    Live v

    This cied,genes,thasetranspenoughabortiis to isettesexpresof Hersine dThe auwhereensureber ofuse ofeffectresistaof liveceptabthe vad dog assays [97,98] (Table 1).st striking aspect of the rst-generation humanthat a leishmanin skin test (LST) is used for

    selection and for conrmation of immunogenic-91]. Whenever the LST is performed, VE is

    mong the individuals whose skin tested positive, whereas no efcacy is detected in assays thatlude an LST [87]. One exception is a study done[88] that showed positive LST but not efcacy,

    ue to the use of a lower number of vaccine dosesn the other hand, a study in Sudan that achievedE against VL is impressive considering the highnd virulence of kala-azar there [91].t-generation vaccines were also used againsteral leishmaniasis (CVL) inducing protection int failing to do so in Brazil [98] (Table 1). Resultsrials of rst-generation human vaccines are also92,99], and they show that immunogenicity isfter a single vaccine dose [92] (Table 1). Further-e vaccines have been used for immunotherapyan CL with success in Brazil [96] and Venezuela

    e 1). Although differing in the use of BCG adju-the number of doses, the authors of both studiesthe vaccine should not be used as monother-tead, combined with the usual chemotherapy, induce toxic and very painful antimonial treatmentased on these results, a rst-generation vaccinegistered as adjunct to antimony therapy in Brazil

    es for second-generation vaccines

    ines

    ory includes vaccines made of genetically mod-ck-out Leishmania spp., which lack essentialh as dyhydrofolate-reductase thymidilate syn-], cystein-proteinase [102,103] or biopterin[104]. These parasites undergo a short life cycle,generate a specic immune response causingfection and no disease in man. Another approachduce in the Leishmania genome suicidal cas-cluding drug sensitive genes, such as L. majorthe ganciclovir-sensitive thymidine kinase genevirus [105] or the Saccharomyces cerevisae cyto-inase gene sensitive to 5-uorocytosine [106].s of these studies have suggested that in Iran,ishmanization is used as vaccine challenge tosistency of infection rates and reduce the num-icipants and duration of the assay [5,106], thesites with suicidal cassettes would guaranteereatment of non-resolving lesions or of infectionsthe usual chemotherapy [106]. However, the usellenge for humans is considered ethically unac-107], and an articial challenge cannot providele information obtained by exposure to natural

  • Vaccines for leishmaniasis in the fore coming 25 years 1713

    Table 1 Vaccine efcacies of rst-generation vaccines (since 1941)

    Disease N Leishmania Country Adjuvant Positive LST (%) VE (%) Treatment Doses Ref.

    CL 1312 Five killed Brazil none 51.5 67.3 Prophylaxis 3 [86]CLCLCLCLVL

    CVLCVL

    CLCL

    CLCL

    infection,saliva [108ticipants nvaccine efcines withones formu

    Vaccinesdelivery v

    Another aplive recomparasite ancarrier andpractical aL. major Ggen, clonedBCG [110];agellar prtid) antige[112]. Examexpressingwhich protexpressingto the recin prime b[114] and d

    Vaccinesantigens

    A further athe puriephoglycanbut becausnever advrst PhaseDunan et apreparatioachieved acinated gro

    ffect candog

    ls: t20] vonov[12

    rmulIIIIil [1mun

    rialsand3%)R. Non wpondn theyearlaceng toThisitan

    easeL-saandoth2597 ALA Columbia none Nd1506 ALA Ecuador BCG 74.43637 ALM Iran BCG 9.852543 ALM Iran BCG 36.22306 ALM Sudan BCG 30

    349 ALM Iran AlOH+BCG Nd1763 Lb Brazil BCG Nd

    900 ALA Venezuela BCG +/ 36.536 ALM Sudan BCG 61.6

    542 Five killed Brazil Nd94 Live and killed Venezuela BCG Nd

    which is modulated by components of sand y]. On the other hand, the large number of par-eeded for a eld trial, due to the low expectedcacy [17], can be reduced only by the use of vac-higher efcacies [17], such as second-generationlated with potent adjuvants.

    using recombinant viruses and bacteria asehicles

    proach to second-generation vaccines is to usebinant bacteria or virus expressing Leishmaniatigen; the bacteria and virus serve as expressionadjuvant system. These vaccines have limited

    pplication. Examples of bacteria vaccines are:P63 surface protease, a major Leishmania anti-in Salmonella thypymurium mutant [7,109] or inthe LCR1 L. chagasi antigen (similar to a T. cruziotein) in BCG [111] and the KMP-11 (kinetoplas-n in attenuated tachyzoites of Toxoplasma gondiiples of vaccines based on virus are Vaccinia virusthe G46/M-2/PSA-2 promastigote surface proteinects against L. amazonensis [113]; or Vacciniathe L infantum LACK antigen (parasite analogueeptor for activated mammalian kinase C) whichoost vaccination, protects mice against L. major

    while eagains

    TwoIII tria[119,1of L. dligand)was foPhasein Brazand imeld t4 obitsdogs (3and PCinfecticorres[11]. Iand 2(25%) pspondi[118].concomthe disthe FMdisease

    The

    ogs against L. infantum infection [64].

    based on puried Leishmania

    pproach to second-generation vaccines includesd Leishmania sub-fractions. Proteins or lipophos-have been used to assess their immunogenicity,e of difculties in their mass production, theyanced to Phase IIa or Phase III trials. In theIII trial with a second-generation dog vaccine,l. [115], using a semi-puried lyophilized proteinn from L. infantum (9467 kDa), paradoxicallysignicantly higher rate of infection in the vac-up than in the control group. This vaccine then,

    of the 54 kprotected dtion. Parasplacebo trvaccinatedfurther perdogs in Souof infectio(12/175) indog showinwas conrmaspirates csis [120]. I[117,118],tion [120],0 Prophylaxis 3 [87]0 Prophylaxis 2 [88]56.7 Prophylaxis 1 [89]35.5 Prophylaxis 1 [90]43.3 Prophylaxis 2 [91]

    69.3 Prophylaxis 1 [97]0 Prophylaxis 3 [98]

    nd Phase I 3 [99]nd Phase I 1 [92]

    76.0 Immunotherapy Monthly [96]95.7 Immunotherapy 3 [100]

    tive in murine models, did not induce protectionine kala-azar in the eld [115].vaccines achieved successful results in Phase

    he FML-saponin [116118] and the LiESAp-MDPaccines. The glycoproteic enriched preparationani promastigotes, named FML (FucoseMannose], antigenic for human [121] and dogs [122],ated with Quillaja saponaria saponin and passedtrials to became the Leishmune licensed vaccine23]. FML was immunogenic, immunoprophylacticotherapeutic, in mice and hamsters, and dogs[116118,124126]. In the rst Phase III assay,6 symptomatic cases among 30 placebo treated

    were detected and conrmed by parasite analysiso obits were detected among vaccines (n = 36) andas conrmed in 3 olygosymptomatic dogs (8.33%)ing to 92% of protection and 76% vaccine efcacysecond assay, the infective pressure was higher

    s after vaccination, obits were detected in 8/33bo treated and 1/20 (5%) vaccinated dogs, corre-95% of protective effect and 80% vaccine efcacyprotection lasted for at least 3.5 years and wast with the reduction of the human incidence ofin the area [118]. Noteworthy, the VE values forponin vaccine showed protection against severeobits due to visceral leishmaniasis.

    er second-generation vaccine, LiESAp, composed

    Da excreted protein of L. infantum plus MDP,ogs in a kennel assay against L. infantum infec-ites were detected in the bone marrow of 3/3eated controls, while they were absent in 0/3dogs [119]. A double-blind randomized trial wasformed with LiESAp +MDP in naturally exposedthern France [120]. After 2 years, the incidencen was 0.61% (1/165) in vaccinees versus 6.86%control dogs, corresponding to a 92% VE. In anyg clinical and/or serological evidence, infectioned by the presence of parasites in bone marrowultured in NNN media and also by PCR analy-n contrast to results of the FML-saponin vaccineLiESAp vaccine induced protection against infec-but not against severe disease or death by VL.

  • 1714 C.B. Palatnik-de-Sousa

    No obits at all were described in the 2 years LiESAp assay[120], reecting the lower infective pressure of the endemicregion. As explained by WHO guidelines [17], conrmationof infection by very sensitive method such as PCR or culture[119,120] represents a very early end-point of infection,while kala-distant endcacies of twinfective pspite of thoage of VE o(IC 95%, 71

    The FMLcine candion Second-in May 20centration,[66] or expgasi. The sulevels inthat the FMThe adjuvand aldehysaponariarst seconveterinarydogs vaccitious, as inof skin-parand lymphblocking vaprotection,80% inhibitmastigote bof in vivo sthe epidemerable, safdogs assays

    Recentlwas elucidattached ta hydrophattached rto the indresponse,C4 is invoicking theprotectiveprotectionto the actresponse atdendritic ctors therebchagasi [13

    Recombi

    The lastthe usesively testrecombina

    [7,16,109,110,133142], in combination [143145], or aspolyproteins or chimeras [65,144,146151]. In order todevelop protection, most of them needed to be formulatedwith adjuvants [17,65,135,137141,143145,147151], ordelivered by bacteria [7,109,110,142,146], with the

    ionmosogen09,1

    advaaysnicas Phseds (Tacondof pin mosursideor D

    itedted csultssizeredtimele.

    says8.32inteity bA (tindugainnt ILromormproeishith M[14

    ime,gasit Lessiondogsith 1d toeers

    Peish1hsp8icalts wted1 (hbinaericntse hiotecazar obits and severe clinical cases [117,118] are-points of infection. A comparison between ef-o vaccines should only be made using the same

    ressure and the same end-point targets [17]. Inse differences, it is worth to note that the aver-f second-generation vaccines together is 82.67%.1394.21).-vaccine is considered a second-generation vac-date and it was featured at the fourth MeetingGeneration Leishmania vaccines held in Merida01 [127]. When used at double adjuvant con-it is also immunotherapeutic for dogs naturallyerimentally infected [63] with Leishmania cha-stained proportions of CD4 and CD21 lymphocyteblood of vaccinated animals [63,66] indicateL-vaccine reduces dog infectivity to sand ies.

    ants used in the FML-vaccine are the QS21de-containing deacylated saponins of Quillaja

    [128,129]. In 2004, the FML-vaccine became thed-generation vaccine licensed for prophylacticuse, under the name of Leishmune [55]. Exposednated with Leishmune proved to be not infec-dicated by the complete absence of clinical signs,asites based on the negative PCR results of bloodnode samples [55]. Leishmune is a transmissionccine [130] for the IgG2 antibodies associated togenerated in vaccinated dogs, [59] caused an

    ion of the in vitro L. donovani and L. chagasi pro-inding to sand y midguts and a 79.3% inhibitionand y infection, helping in the interruption ofics [130]. Leishmune vaccine proved to be tol-e, and highly immunogenic [123] in a recent 600in the eld.

    y, the mechanism of action of the QS21 saponinated. QS21 contains two carbohydrate chainso a triterpene nucleus C3 and C28, besidesobic moiety which is acylated to a C28 sugaresidue. The QS21 hydrophobic moiety is relateduction of the CTL CD8+ protective lymphocytewhile the aldehyde group present in triterpenelved in direct T lymphocyte stimulation, mim-B7-1 co-stimulatory molecule to induce the TH1response [131]. Preliminary results suggest thatinduced by the FML-QS21 vaccine is also relatedivation of a bradikinin mediated inammatorythe site of injection, which stimulates immatureells through their B1R and B2R surface recep-y triggering a TH1 response against Leishmania2].

    nant antigens

    approach in second-generation vaccines isof recombinant proteins that were inten-ed since the 1990s (Table 2). The Leishmaniant vaccine candidates were assayed alone

    exceptWhileimmun[7,17,1themnel asspre-clito dogwere uspecieand seresultslationno expwe conbinantof limuntreathe reof thecompalatestavailabthe as95%, 5

    AnimmunThe TSstresskeys aadjuvamay plong-tefusionLeIF (Ltion wand VLAdjuPrL. chaprevenprogreThesecine wshowevoluntand inthe Le83 (Lbto clinpatienprotecHASPBin coma chimfragmeand thtial prof LeIF [134] and HASPB1 proteins [133].t recombinant proteins were assayed for theiricity and protective potential in mice models10,133137,139143,149,150], only a few ofnced to monkey trials for CL [143,138], to ken-in dog model against VL [65,144146,151], or tol studies in humans [147,148]. None has advancedase III trials (Table 2). The recombinant proteinsagainst all forms of the disease and all parasiteble 2). In contrast to the results for rst (Table 1)-generation vaccines with native antigens, therotection developed by each recombinant formu-ice do not allow determination of VE values, sincee to natural infection occurred. In this review,red for comparison of protection due to recom-NA vaccines, the reported reduction of LDU ordilution values in vaccinated mice compared toontrols. In the specic case of CL mouse models,were obtained from each report as the percentof the footpad lesion of the vaccinated animalsto that of the untreated saline control, at thepoint when data for the saline control was still

    The mean average of parasite reduction for allwith recombinant antigens (Table 2) is 68.02% (IC77.71).resting approach is the induction of protectivey a polyprotein vaccine formulation (Table 2).hiol-specic antioxidant) and LmSTI1 (L. majorcible protein 1) are protective for mice and mon-st CL [143], although the use of recombinant-12 is not recommended at present because itte immune disorders [152] and fail to induce

    immunity [135]. The multicomponent Leish-111ftein containing the antigens TSA, LmSTI1 andmania elongation initiation factor), in formula-PL-SE9 and squalene, protect mice against CL9,150,153] but, in combination with MPL-SE orwas only immunogenic in dogs challenged with

    [65] and L. infantum (MML) [144], and failed toishmania infantum natural infection, or diseasein dogs in an open kennel trial [151] (Table 2).[151] received two courses of three-dose vac-year interval. Nevertheless, Leish111f +MPL-SE

    be safe, immunogenic, and reactogenic in healthyin the US and in patients of CL and ML in Brazilru, respectively [148], while vaccination with11f components, Leishmania heat shock protein3) and GM-CSF, combined to chemotherapy, ledimprovement and complete cure of six humanith MCL [147]. Furthermore, the H1 histone thatmice [137] and monkeys against CL [138], theydrophilic acylated surface protein B1) or bothtion with Montanide [144], and the protein Q,antigen composed of the genetic fusion of veof the acidic ribosomal protein Lip2a, Lip2b, P0stone H2A used with BCG [146] developed par-tion against CVL in dogs against infection and at

  • Vaccinesfor

    leishmaniasis

    inthe

    forecom

    ing25

    years1715

    Table 2 Second-generation recombinant antigens (1990s)

    Antigen Adj./delivery system Model Disease Leishmania Parasite reduction (%) Phase Ref.

    GP63 S. thyphimurium Mice CL L major 6778 IIIa [7,109]GP63 BCG Mice CL L. mexicana 6883 IIIa [110]GP63 BCG Mice CL L. major 42 IIIa [110]HASPB1 Mice CL L. donovani 60 IIIa [133]LeIF Mice CL L. major 57 IIIa [134]TSA and LmSTI1 IL12 Mice CL L. major 9899 IIIa [143]

    IL12 and alum Monkey CL L. major 97 IIa [143]Leish 111 = LeIF + TSA + LmSTI1 MPL-SE/RIBI 529-SE Mice CL L. major 5367 IIIa [149]Leish 111 = LeIF + TSA + LmSTI1 MPL-SE/Adjuprime Mice VL L. infantum 99.6 IIIa [150]Leish 111 = LeIF + TSA + LmSTI1 MPL-SE/Adjuprime Dog CVL L. chagasi I [65]Leish 111 = LeIF + TSA + LmSTI1 MPL-SE/Adjuprime Dog CVL L. infantum Open kennel [151]MML MPL-SE Dog CVL L. infantum IIa [144]LeIF + TSA + LmSTI1 + Lbhsp83 GM-CSF Human MCL L brasiliensis Cure ita [147]Leish 111 = LeIF + TSA + LmSTI1 MPL-SE Human CL Safe I [148]LACK IL-12 Mice CL L. major 53 IIIa [135]LACK Mice CL L. amazonensis 0 IIIa [136]H1 IFA and IL-12 Mice CL L. major 6453 IIIa [137]H1 Montanide Monkey CL L. major 37.5 IIa [138]H1, HASPB1, H1 +HASPB1 Montanide Dog CVL L. infantum 5/84/8 IIa [144]CPb Poloxamer 407 Mice CL L. major. 41 IIIa [139]CPb +CPa QuilA + IL-12 Dog CVL L. infantum 0 IIIa [145]PSA-2 C. parvum/ISCOMs Mice CL L. major 0 IIIa [140]LCR1 CFA Mice VL L. chagasi 67 IIIa [141]NH36 Q. saponaria saponin Mice VL L. chagasi 79 IIIa [17]A2 P. acnes Mice VL L. donovani 88 IIIa [142]Protein Q chimera BCG Dog CVL L. infantum 5090 IIIa [146]a it = Immunotherapy.

  • 1716C.B.

    Palatnik-de-Sousa

    Table 3 Candidates for third-generation-DNA vaccines against leishmaniasis (1990s)

    Antigen Adj./delivery system Model Disease Leishmania g Virulence Challenge Parasitereduction (%)

    Phase Ref.

    GP63 pCMV Mice CL L. major 100 NV 1 106 pro 57 IIIa [14]LACK pcDNA3 Mice CL L. major 100 NV 1 105 pro 42 IIIa [135]LACK pcDNA3 Mice CL L. major 100 NV 1 105 pro 56 IIIa [9]LACK pcDNA3 IL-12 Mice VL L. donovani 100 NV 1 106 pro 0 IIIa [162]HPB-LACKa pcDNA3-Vaccinia Virus Mice CL L. major 100 NV 5 104 pro 68 IIIa [114]HPB-LACKa pCIneo-Vaccinia Virus Dogs CVL L. infantum 100 V 1 108 pro 3/5 IIIa [64]LACK pCMV3ISS Mice CL L. major 50 V 5 103 ama 26 IIIa [156]GP63 pCMV3ISS Mice CL L. major 50 V 5 103 ama 21 IIIaP20 pCMV3ISS Mice CL L. major 50 V 5 103 ama 20 IIIaPSA2 pCMV3ISS Mice CL L. major 50 V 5 103 ama 26 IIIaTSA pcDNA3 Mice CL L. major 100 V 1 104 ama 84 IIIa [157]LmSTI1 pcDNA3 Mice CL L. major 100 V 1 104 ama 32 IIIaTSA + LmSTI1 pcDNA3 Mice CL L. major 100 V 1 104 ama 90 IIIaH2A +H2B +H3 +H4 pcDNA3 Mice CL L. major 200 NV 5 104 pro 74 IIIa [158]Cpa +CPb pCB6 Mice CL L. major 200 NV 1 106 pro 38 IIIa [159]HPB CPa and CPba pCB6+ Montanide 720

    and CPGDog CVL L. infantum 200 NV 5 106 pro 8/10 IIIa [15]

    KMPII, TRYP, LACK and GP63 pMOK Dog CVL L. infantum 800 V 5 107 pro 0/12 IIIa [51]P4 pcDNA3 Mice CL L. amazonensis 100 V 2 105 pro 25 IIIa [168]P4 pcDNA3 +HSP70 Mice CL L. amazonensis 100 V 2 105 pro 60 IIIaP4 pcDNA3 + IL-12 Mice CL L. amazonensis 100 V 2 105 pro 99 IIIaA2 pcDNA3 Mice VL L. donovani 100 NV 2 108 pro 97 IIIa [160]ORFF pcDNA3 Mice VL L. donovani 100 NV 2 108 pro 59 IIIa [163]NH36 VR1012 Mice VL L. chagasi 100 V 2 108 ama 88 IIIa [16]NH36 VR1012 Mice CL L. mexicana 100 V 1 106 pro 65 IIIa [16]NH36 VR1012 Mice CL L. amazonensis 100 V 1 106 pro 81 IIIa [164]NH36 VR1012 Dog VL L. chagasi 750 CVL 7 108 ama 4/6 IIIa [165]KMP11 pCMV-LICK Hamster VL L. donovani 100 V 1 106 pro 85 IIIa [166]papLe22 pcDNA3.1 Hamster VL L. infantum 100 V 1 107 ama 50 IIIa [161]NH36 VR1012 Mice VL L. chagasi 100 V 2 108 ama 91 IIIa [167]a HPB =Heterologous prime-boost.

  • Vaccines for leishmaniasis in the fore coming 25 years 1717

    the clinical level. Finally, poloxamer 407 adjuvant CPb, butnot CPa was able to protect mice against CL [139] whereasboth in combination with IL-12 and QuilA, failed to protectdogs from L. infantum infection, probably due to the lowconcentration of the adjuvants (50g in each vaccine dose)[145].

    As singlethe L. maplasmid DNnant PSA2or with C.tion againsthe amastition to VL.an essentianucleoside[16], sincebiosynthescomplex [1CVL [55]. Tlaja saponchagasi anresponse [1

    In 1996studies ofthat time.Leishmaniatrends in Leantigens intion in navprotectedNo assays wof reductioproblem wtransportasevere procine produ

    Candidat

    Comparedare much mcost of proand exibistruct. Thepotent immtion of innasequencesthe host, leteins for lo

    The moviously as137,1561[9,14,16,13of genes(HPB), whfollowed[15] or atein [64,1in only twobserved in

    [14,16,64,135,156,159,161,164167], with the exceptionof pMOK in a dog assay [51], and of pcDNA3 in a miceassay [162]. Most trials were performed in mice againstCL and VL [9,14,16,114,135,156160,162168], and somein the hamster against VL [166,161] and in dogs against

    5,51,64,165]. All the studies involved articial chal-(Phase IIIa) with agents of the New and Old Worldaniasis (Table 3). The most striking characteristic ofA vaccine studies is that about a half of them usedallenge non-virulent (NV) Leishmania strains, grownid culture media [9,14,15,114,135,158160,162,163]a low number of parasites [9,114,135,158], while

    st used virulent [V] parasites isolated from infecteds [16,51,64,156,157,161,164168] (Table 3). Thisgeneity of protocols determined a high variation ofts of reduction of parasite load, with a mean average4% (IC 95% 47.7570.73).diffdifresuthis

    pe of2 [16isogusedue

    otecof Ne ney.vac

    se Isedy, avelo

    s goiLeIF,mo

    rth cesteice,

    tedni [A2 ginfe3) p4]. TIL-12o Ig

    4nes

    idate

    TSA,

    CPb1candidate recombinant vaccines tested in mice,jor LACK with IL-12 was less efcient than theA encoding LACK [135,136,154], the recombi-antigen (parasite surface antigen 2) in ISCOMsparvum, induced a TH1 response but not protec-t CL [136,140], while the LCR1 protein [141] andgote-specic A2 antigen [142] conferred protec-The nucleoside hydrolase of L. donovani (NH36) isl enzyme that releases DNA bases from importeds, allowing the parasite to construct its own DNALeishmania parasites lack the de novo purine

    is pathway. NH36 is the main antigen of the FML55] that constitutes the Leishmune vaccine forhe recombinant NH36 in combination with Quil-aria Molina saponin protected mice against L.d L. mexicana infection (Table 2) through a TH16]., the WHO-TDR program organized comparativeseveral leading recombinant proteins known atIn 2001, the fourth Meeting on Second-Generationvaccines evaluated these studies and discussedishmania vaccine development [127]. While mostduced lymphocyte proliferation and IFN secre-emice and human patients, the only antigens thatmice against CL were the MIX, LACK, 4H6 and FPA.ere performed against VL and no quanticationn of parasitic load was done. The main detectedas the lack of stability and potency during thetion of the antigens [127], suggesting potentiallyblems with future scale-up of recombinant vac-ction.

    es for third-generation vaccines

    to recombinant protein vaccines, DNA vaccinesore stable and have the advantage of their lowduction, no need of cold chain for distribution,lity of combining multiple genes in a simple con-mechanism by which DNA vaccination generatesune responses appears to be through the activa-te immune responses by the non-methylated CpGof bacteria and to the intense replication withinading to the expression of the recombinant pro-nger periods.st-studied antigens (Table 3) were those pre-sayed as recombinant proteins [14,16,51,135,61]. Most of them were tested as single vaccines5,156,157,160167], and some, as combination

    [156159,168] or as heterologous prime-boostich involves an injection of the DNA vaccineby an injection of the recombinant proteinVaccinia virus expressing the recombinant pro-14]. Adjuvants were added to formulationso studies [15,162] (Table 3). Protection wasvaccines using all the tested plasmids (Table 3)

    CVL [1lengeleishmthe DNfor chin liquand atthe reanimalheteropercenof 59.2

    Thefor theversialrefutethe tyof IL-1tion inthe motectionand pr750ggest thefcac

    DNAno Phais focuefcaccine destudieLACK,are thethe fobeen t

    In mprotecdonovaand PSmajor(TableCVL [6 andIgG2 t

    Tablevacci

    Cand

    LACKLeIF,H1Cpa +KMP1NH36erent codied antigens could be the responsibleferent degrees of protection. However, contro-lts for the same antigen [9,64,114,135,156,162]idea (Table 3). Protection did not correlate toexpression vector (p > 0.05), or to the addition2,168]. It seems to be easier to achieve protec-enic mice than in dogs [15,51,64,165]. Most ofinvestigations used 100g of plasmid, while pro-to CP proteinase vaccine required 200g [159],

    tion of dogs against CVL was achieved by usingH36 plasmid DNA vaccine [165]. These results sug-ed for higher plasmid concentrations to achieve

    cines are indeed protective, although to dateII trial data are available. Although this reviewon vaccine candidates that already show anylot of interest was though generated in vac-pment against leishmaniasis in recent years, withng on in the labs on experimental models. TheTSA, LmSTI1, H1, CpA +CpB, KMP11 and NH36

    st promising candidates that may nd a place inoming years (Table 4), since they have alreadyd in more animal models.LACK DNA induced a TH1 response [9,156] that

    against infection by L. major [9,135], but not L.162]. Even truncated portions of the LACK geneene were superior to GP63 and p20 against L.ction [156], and immunization with HPB-LACKrotected mice against VL [114] and dogs fromhe immunization resulted in an increase in IFN-expression, lymphocyte proliferative response,

    G1 ratio while it led to decreases in clinical

    Promising candidates for third-generation

    antigen Tested animal model

    Mice and dogsLmSTI1 Mice, monkey, human

    Mice, monkeyMice, dogsMice, hamsters, dogsMice, dogs

  • 1718 C.B. Palatnik-de-Sousa

    Table 5 Efcacy of vaccines for leishmaniasis developed so far (19412007)

    Generation Adjuvant Phase Average protection IC 95% Evaluation Model

    1st Yes/no III 54.38 39.8468.92 VE Human, dog2nd native 71.1294.20 VE Dog2nd recom 58.3277.71 Parasite load Mice, monkey, dog3rd 47.7

    symptoms,expression

    VaccinaDNA vaccin[157], protThe digenethe latter i

    InjectioH3 and H4)infection t

    While pcombinatiomajor [159720 and CPincrease inliferation,or clinical slow infectivaccinatedPCR negatision couldtwo untrea

    Vaccinaagainst VL[166], whilLACK, anddogs agains

    The NH3by L. chagaindicatingprophylactProtectionby the recoand it is rare characdog Phasethe NH36 vobserved soapeutic PSAskin lesionparasite lochagasi [16

    The comferent gensummarizewith or wisecond-gensignicantthese twoarise fromthe target

    the-genbinam t

    sultsion oudie

    nes

    onces tesigns thbilitcase15 ad su[175

    eti

    repplayy epsyntas sspechrouproaes ag

    lusi

    e ofw knP ree pof scieies,creasaniaYes III 82.66binant Yes IIa 68.02

    No IIa 59.24

    number of parasites in target tissues, and IL-4.tion of mice either with the TSA or the LmSTI1es, or with both as a tandem digene constructected against CL through a CD4 +TH1 response.and the TSA gene were the most protective, withnvolving a CD8+ response [157].n of a mixture of four histone plasmids (H2A, H2B,in Balb/c mice also protected against L. major

    hrough a TH1 response [158].lasmid CPa and CPb on their own did not, then of both conferred protection on mice against L.], and a prime boost vaccination with MontanideG protected dogs against CVL, as evidenced by theIgG2 specic antibody synthesis, lymphocyte pro-IFN/IL10 secretion, and DTH response. No deathigns were reported [15], probably due to the veryve challenge (5 106 promastigotes). Eight of 10dogs were considered protected based on theirve results [15]; however, no signicant conclu-be drawn from the study, because it used onlyted control dogs.tion with 200g of KMP11 protected hamstersthrough a mixed cytokine TH1/TH2 response

    e a cocktail of plasmid DNA encoding KMPII, TRYP,GP63 (200g of each plasmid) did not protectt L. infantum virulent challenge [51].6 DNA vaccine protected mice against infectionsi, L. mexicana [16], and L. amazonensis [164],its potential usefulness in a bivalent immuno-ic vaccine for the control of both endemics.by the DNA vaccine was higher than that inducedmbinant NH36 or the FML antigen plus saponin,elated to IFN--producing CD4+ T cells, whichteristic of a TH1 type immune response [16]. AI-IIa study on the immunoprophylactic effect ofaccine is in progress, and protection has beenfar in 4/6 vaccinated dogs [165]. While the ther-/GP46 DNA vaccine caused reduction of L. major

    s in mice [169], the NH36 DNA vaccine reducesad and increases survival of mice infected with L.7].parison of the efcacies achieved by the dif-erations of vaccines against leishmaniasis isd in Table 5. While the rst-generation vaccines,

    whilesecondrecoment frothe rereducttory st

    Vacci

    Basedenhanbeen dantigethe viais thethe SPinducemajor

    Synth

    Recentmightidentiffuturetein wwhichcells tthis apvaccin

    Conc

    In spitthe slothe GMest, thlack ocountrthe inleishmthout adjuvant, display a relatively low VE, theeration vaccines with native antigens show aincrease in VE. The VE results derived fromgroups of vaccines are very robust, since theyeld trials with exposure to natural challenge inspecies (human and dogs). On the other hand,

    ous use ofdelay in thHealth. Ththe fore cocomposedadjuvants.570.73 Parasite load Mice, dog

    protection induced by the recombinant proteineration vaccines are slightly greater, neither thent nor the DNA vaccines are signicantly differ-he rst-generation ones. It is worth noting thatof these last two groups are calculated based onf parasite load achieved mainly in mice labora-s.

    based on sand y salivary antigens

    the observations that the saliva of sand ieshe infectivity of pathogens [170], vaccines havened against components of saliva or insect gutat can protect from infection [171] and decreasey and reproducibility of the insect [172,173]. Thisof the protein MAX or MAXADILAN [174] and of

    ntigen obtained from Phlebotomus papatasi thatbstantial resistance in mice to infection by L.].

    c vaccines

    orts suggest that CD8+ in addition to CD4+ T cellsa role in defence and cure of leishmaniasis. Toitopes recognized by CD8+ T cells to be used inhetic vaccines, the sequence of the KMP11 pro-canned and thirty nonapeptides were identiedically trigger IFN- secretion by human CD8+gh the MHC class system [176]. In the future,ch will allow the development of synthetic T cellainst leishmaniasis.

    ons and perspectives

    the many genes identied as vaccine candidates,owledge transfer from the laboratory to industry,gulations that dampen down the industrial inter-orly developed biotechnology industry and thentists in regulatory agencies of underdevelopedthe ethical constraints on research in animals anding dog-chemotherapy in Europe, where humansis is less frequent, contribute to the continu-

    rst-generation and live vaccines, and to thee arrival of combined DNA vaccines to Publice prediction is that vaccines for leishmaniasis inming 25 years will be of second-generation type,of complex native antigens and well developed

  • Vaccines for leishmaniasis in the fore coming 25 years 1719

    Acknowledgements

    This work was nancially supported by Conselho Nacionalde Desenvolvimento Cientco e Tecnologico (CNPq) (Edi-tal Universal and productitivity fellowship), Fundacaode Ampar(FAPERJ-PRFAPERJ-INF

    Referenc

    [1] WorldgrammImpro9900/

    [2] Tesh Rto cha

    [3] AlvarF, Lopciency1997;1

    [4] GavroIsrael:

    [5] KamesLeishmovervi

    [6] Modabasitol

    [7] YangLP, Liexpreerentiagains

    [8] SkeikyKubinulatesa Th1Exp M

    [9] GurunGlaichimmuimmu1997;1

    [10] JardimLeishmtive cImmu

    [11] Rachavanileishm

    [12] PalatnInhibition inglycoc

    [13] Xu D,cine 1

    [14] Liew Fof DNmajor

    [15] Rafatiet al.niasiswith cVaccin

    [16] AguilaGP, Ro

    efcacy of a prophylactic Leishmania donovani DNA vaccineagainst visceral and cutaneous murine leishmaniasis. InfectImmun 2005;73:8129.

    [17] UNDP/World Bank/WHO Special Programme for Researchand Traininbg in Tropical Diseases (TDR). Guidelines for thevaluaions eealtheinero Le7.elkaiD25+enceempaniaaratt, Maruringance.acelle Jeespon002;7halibassanatholvestharazA, etett 1nsariammiseaseruhy, Rabory/rf inteumouctiveasi ine Luiagolacebcandiasis.otoxperi004;3awndereion inicroboolerishmeruhy, Rabhum

    etweome.asaktudieell-mivtymun

    odrigactorishmo a` Pesquisa do Estado do Rio de JaneiroONEX; CNE fellowship; Edital Pensa Rio; EditalRA).

    es

    Health Organization-TDR. The TDR fteenth pro-report. Research Progress 19992000. New and

    ved Tools; 2003. www.who.int/tdr/research/progresstools/vdr.htm.. Control of zoonotic visceral leishmaniasis. Is it timenge strategies? Am J Trop Med Hyg 1995;52:28792.J, Canavate C, Gutierrez-Solar B, Jimenez M, Lagunaez-Velez R, et al. Leishmania and human immunode-virus coinfection: the rst 10 years. Clin Microbiol Rev0:298319.n D, Saul A. Pioneer of tropical medicine. Rehovot,Balaban Publishers; 1997, ISBN 0-86689-045-9.ipour A, Rafati S, Davoudi N, Maboudi F, Modabber F.aniasis vaccine candidates for development: a globalew. Ind J med Res 2006;123:42338.ber F. Vaccines against leishmaniasis. Ann Trop Med Par-1995;89:838.DM, Fairweather N, Button LL, McMaster WR, Kahlew FY. Oral Salmonella typhimurium (AroA-) vaccinessing a major leishmanial surface protein (gp63) pref-ally induces T helper 1 cells and protective immunityt leishmaniasis. J Immunol 1990;145:22815.YAW, Guderian JA, Benson DR, Bacelar O, Carvalho EM,M, et al. A recombinant Leishmania antigen that stim-human peripheral blood mononuclear cells to express

    -type cytokine prole and to produce interleukin 12. Jed 1995;181:152737.athan S, Sacks DL, Brown DR, Reiner SL, Charest H,enhaus N, et al. Vaccination with DNA encoding thenodominant LACK parasite antigen confers protectivenity to mice infected with Leishmania major. J Exp Med86:113747.A, Tolson DL, Turco SJ, Pearson TW, Olafson RW. Theania donovani lipophosphoglycan T lymphocyte reac-

    omponent is a tightly associated protein complex. Jnol 1991;147:353844.mim N, Jaffe CL. Pure protein from Leishmania dono-protects mice against both cutaneous and visceralaniasis. J Immunol 1993;150:232231.ik CB, Borojevic R, Previato JO, Mendonca-Previato L.tion of Leishmania donovani promastigote internaliza-to murine macrophages by chemically dened parasiteonjugate. Infect Immun 1989;57:75463.Liew FY. Genetic vaccination against Leishmaniasis. Vac-994;12:15346.Y, Xu D. Protection against leishmaniasis by injectionA encoding a major surface glycoprotein, gp63, of L.. Immunology 1995;84:1736.S, Nakhaee A, Taheri T, Taslimi Y, Darabi H, Eravani D,Protective vaccination agaisnt canine visceral leishma-using a combination of DNA and protein immunizationysteine proteinases typeI and Type II of L. infantum.e 2005;23:371625.r-Be I, Zardo RS, Paraguai de Souza E, Borja-Cabrerasado-Vallado M, Mut-Martin M, et al. Cross-protective

    etH

    [18] Rt7

    [19] BCt

    [20] Km

    [21] BWdt

    [22] Bdr2

    [23] GHpIn

    [24] KJL

    [25] Agd

    [26] PAtotag

    [27] Dtpan

    [28] Ge2

    [29] HAsM

    [30] Cle

    [31] PAinbc

    [32] BscitIm

    [33] Rfletion of Plasmodium falciparum vaccines in popula-xposed to natural infections. TDR/MAL/VAC/97. Worldorganization, Geneva, Switzerland.SL, Locksley RM. The regulation of immunity

    ishmania major. Annu Rev Immunol 1995;13:151

    d Y, Piccirillo CA, Mendez S, Shevach EM, Sacks DL. CD4+regulatory T cells control Leishmania major persis-

    and immunity. Nature 2002;420:5027.K. Cytokine producing T cells subsets in human leish-sis. Arch Immunol Ther Exp (Warz) 2000;48:1736.a-Masini A, Teixeira-Carvalho A, Malaquias LC, Mayrinktins-Filho OA, Correa-Oliveira R. Mixed cytokine proleactive cutaneous leishmaniasis and in natural resis-Front Biosci 2007;12:83949.

    ar O, Lessa H, Schriefer A, Machado P, Ribeirosus A, Dutra WO, et al. Up-regulation of Th1-typeses in mucosal leishmaniasis patients. Infect Immun0:673440.HW, Piuvezam MR, Skeiky YA, Siddig M, Hashim FA, el-AM, et al. Interleukin 10 production correlates with

    ogy in human Leishmania donovani infections. J Clin1993;92:3249.mi A, Kemp K, Ismail A, Gasim S, Gaafar A, Kurtzhalsal. T cell response in human leishmaniasis. Immunol

    999;65:1058.NA, Saluja S, Salotra P. Elevated levels of interferon-a, interleukin-10, and interleukin-6 during activee in Indian kala azar. Clin Immunol 2006;119:33945.pe-Magalhaes V, Martins-Filho OA, Prata A, Silva Ldeello A, Teixeira-Carvalho A, et al. Mixed inamma-egulatory cytokine prole marked by simultaneous raiserferon-gamma and interleukin-10 and low frequency ofr necrosis factor-alpha(+) monocytes are hallmarks ofhuman visceral Leishmaniasis due to Leishmania cha-fection. Clin Exp Immunol 2006;146:12432.ca PM, Mayrink W, Pinto JA, Coutinho SG, San-MA, Toledo VP, et al. A randomized double-blindo-controlled trial to evaluate the immunogenicity ofidate vaccine against American tegumentary leishma-Acta Trop 2001;80:25160.

    H, Lindoso JAL. Immunity and immunosuppression inmental visceral leishmaniasis. Braz Med J Biol Res7:61523.TR, Ozinsky A, Underhill DM, Buckenr FS, Akira S,m A. Leishmania major activates IL-1 alpha expres-

    macrophages thorugh a MyD88-dependent pathway.es Infect 2002;4:76371.RN, Reed SG. Second-generation vaccines against

    aniasis. Trends Parasitol 2005;21:2449.pe-Magalhaes V, Martins-Filho OA, Prata A, Silva Ldeello A, Teixeira-Carvalho A, et al. Immune responsean visceral leishmaniasis: analysis of the correlationen innate immunity cytokine prole and disease out-Scand J Immunol 2005;62:48795.SK, Saha B, Battacharaya A, Roy S. Immunobiologicals on experimental visceral leishmaniasis. II. Adherentediated down-regulation of delayed-type hypersensis-response and up-regulation of B-cell activation. Eur Jol 1992;22:20415.ues Jr V, Silva JS, Campos-Neto A. Transformig growth- and immunosupression in experimental visceral

    aniasis. Infect Immun 1998;66:12336.

  • 1720 C.B. Palatnik-de-Sousa

    [34] Gomes NA, Barreto-de-Souza V, Wilson ME, Dos Reis GA.Unresponsiveness CD4+ T lymphocytes from leishmanaichagasi-infected mice increase cytokine production andmediate parasite killing after blockade of B7-1; CTLA-4molecular pathway. J Inf Dis 1998;178:184751.

    [35] Gomeinfect

    [36] Alexanthe eadonov

    [37] VasconSuppreserumrole in

    [38] Ato Mexpresof visc

    [39] JancoduringTrends

    [40] Rogersto Lei

    [41] Gumywith Lof metiationParasi

    [42] Sacksand re2002;2

    [43] Solbaction. A

    [44] AnamRP, etvaluelin G31999;6

    [45] Atta Avalhoactive1998;5

    [46] da Mation oafter tAnal 2

    [47] RyanStitelebasedlin M (cutane

    [48] AnamGoswabraneIgG suchemo

    [49] RavindSK, etclass rchemo

    [50] KeenaAJ. ViInfect1984;2

    [51] RodrgL, Solencoddogs aVaccin

    [52] Almeida MAO, Jesus EEV, Sousa-Atta MLB, Alves LC, BerneMEA, Atta AM. Antileishmanial antibody prole in dogsnaturally infected with Leishmania chagasi. Vet ImmunolImmunopathol 2005;106:1518.

    [53] Iniesta L, Gallego M, Portus M. Immunoglobulin G and Eesponmun

    ourteiousnontrofectogueParrf cansitesogs.eplaznd Ignd otietolonsootalorreleishmolanoadareG2 areas.ent.endeatistodyogs589uinnye Cf can003;9ourdetectmbediagnantanR, Parom LeralantosB, Manineeishmamirstebaaniamun

    NA vACK.ujiwada S,hreeial vaes 20orja-kadamun

    ML-va J,oughespons NA, Dos Reis GA. The dual role of CTLA-4 in Leishmaniaion. Trends parasitol 2001;17:48791.der CE, Kaye PM, Engwerda CR. CD95 is required forrly control of parasite burden in the liver of Leishmaniaani-infected mice. Eur J Immunol 2001;31:1199210.cellos RCS, Urago KP, Bunn-Moreno MM, Madeira ED.ssor activity in Leishmania donovani-infected hamster: reversion by delipidated bovine serum albumin andcycle events. Braz J med Biol Res 1996;29:61522.

    , Stager S, Engwerda CR, Kaye PM. Defective CCR7sion on dendritic cells contributes to the developmenteral leishmaniasis. Nat Immunol 2002;3:118591.vic D, Liu Z, Gause WC. Th1 and Th2 cell commitmentinfectious disease:asymmetry in divergent pathways.Immunol 2001;22:4507.KA, Dekrey GK, Mbow ML. Type I and type 2 responses

    shmania major. FEMS Microbiol Lett 2002;209:17.A, Louis JA, Launois P. The murine model of infectioneishmania major and its importance for the decipheringchanisms underlying differences in Th cell differen-in mice from different genetic backgrounds. Int J

    tol 2004;34:43344.D, Noben-Trauth N. The immunology of susceptibilitysistence to leishmnai major in mice. Nat Rev Immunol:84558.h W, Laskay T. the host response to Leishmania infec-dv Immunol 2000;74:275317.K, Afrin F, Banerjee D, Pramanik N, Guha SK, Goswamial. Immunoglobulin subclass distribution and diagnosticof Leishmania donovani antigen-specic immunoglobu-in Indian kala-azar patients. Clin Diagn Lab Immunol:2315.M, DOliveira, Correa J, Atta ML, Almeida RP, Car-EM. Anti-leishmanial IgE antibodies: a marker ofdisease in visceral leishmaniasis. Am J Trop Med Hyg9:42630.tta VL, Hoshino-Shimizu S, Dietze R, Corbett CE. Detec-f specic antibody isotypes and subtypes before andreatment of American visceral leishmaniasis. J Clin Lab000;14:512.JR, Smithyman AM, Rajasekariah GH, Hochberg L,r JM, Martin SK. Enzyme-linked immunosorbent assayon soluble promastigote antigen detects immunoglobu-IgM) and IgG antibodies in sera from cases of visceral andous leishmaniasis. J Clin Microbiol 2002;40:103743.K, Afrin F, Banerjee D, Pramanik N, Guha SK,mi RP, et al. Differential decline in Leishmania mem-antigen-specic immunoglobulin G (IgG), IgM, IgE, andbclass antibodies in Indian kala-azar patients aftertherapy. Infect Immun 1999;67:66639.ran R, Anam K, Bairagi BC, Saha B, Pramanik N, Guhaal. Characterization of immunoglobulin G and its sub-esponse to Indian kala-azar infection before and aftertherapy. Infect Immun 2004;72:86370.n CM, Hendricks LD, Lightner L, Webster HK, Johnsonsceral leishmaniasis in the German shepherd dog. I.ion, clinical disease, and clinical pathology. Vet Pathol1:749.uez-Cortes A, Ojeda A, Lopez-Fuertes L, Timon M, Atletano-Gallego L, et al. Vaccination with plasmid DNAing KMP11, TRYP, LACK, and GP63 does not protectgainst Leishmania infantum experimental challenge.e, in press.

    rIm

    [54] CtcIn

    [55] NI,oad

    [56] Daa

    [57] NAtcl

    [58] SlIgam

    [59] MBbd2

    [60] QDo2

    [61] BdeD

    [62] SNfc

    [63] SAcL

    [64] REmimDL

    [65] FJttR

    [66] BOimF

    [67] MCrses in various stages of canine leishmaniosis. Vetol Immunopathol 2005;103:7781.nay O, Quinnell RJ, Garcez LM, Shaw JJ, Dye C. Infec-ess in a Cohort of Brazilian dogs: why culling fails tol visceral leishmaniasis in areas of high transmission. JDis 2002;186:131420.ira FS, Moreira MAB, Borja Cabrera GP, Santos FN, Menza LE, et al. Leishmune vaccine blocks the transmissionine visceral leishmaniasis. Absence of Leishmania par-in blood, skin and lymph nodes of vaccinated exposedVaccine 2005;23:480510.es P, Smith NC, Arnold P, Lutz H, Eckert J. Specic IgG1G2 antibody responses of dogs to Leishmania infantumher parasites. Parasite Immunol 1995;17:4518.CG, Garcia Alonso M, Requena JM, Miron C, Soto M,C, et al. Analysis of the humoral response against

    and recombinant antigens of Leishmania infantum:ation with disease progression in canine experimentalaniasis. Vet Immunol Immunopathol 1999;67:11730.-Gallego L, Riera C, Roura X, Inieste L, Gallego M, Val-s JE, et al. Leishmania infantum-specic IgG, IgG1 andntibody responses in healthy and ill dogs from endemicEvolution in the course of infection and after treat-Vet Parasitol 2001;96:26576.s CO, Paraguai de Souza E, Borja-Cabrera GP, Meloa LM, Santos MA, Parra LE, et al. IgG1/IgG2 anti-dichotomy in sera of vaccinated or naturally infectedwith visceral leishmaniasis. Vaccine 2003;21/1920:97.ell RJ, Courtnay O, Gacez LM, Kaye PM, Shaw MA,, et al. IgG subclass responses in longitudinal studyine visceral leishmaniasis. Vet Immunol Immunopathol1:1618.oiseau G, Marchal T, Magnol JP. Immunohistochemicalion of Leishmania infantum in formalin-xed, parafn-ded sections of canine skin and lymph nodes. J VetInvest 1997;9:43940.a DM, Borja-Cabrera GP, Paraguai de Souza E, Sturmlatnik de Sousa CB, Campbell DA. Nucleoside hydrolaseeishmania (L.) donovani is an antigen diagnostic for vis-leishmaniasis. Mol Biochem Parasitol 2002;120:3159.FN, Borja-Cabrera GP, Miyashiro LM, Grechi J, Reis

    oreira MA, et al. Immunotherapy against experimentalvisceral leishmaniasis with the saponin enriched-

    une vaccine. Vaccine 2007;25:617690.o MJ, Zarate JJ, Hanke T, Rodriguez D, Rodriguez JR,n M, et al. Protection in dogs against visceral leish-sis caused by Leishmania infantum is achieved byization with a heterologous prime-boost regime usingaccine and vaccinia recombinant vectors expresssingVaccine 2003;21:247484.ra RT, Vale AM, Franca da Silva JC, da Costa RT, QuetzMartins Filho OA, et al. Immunogenicity in dogs of

    recombinant antigens (TSA, LeIF and LmSTI1) poten-ccine candidates for canine visceral leishmaniasis. Vet05;36:82738.Cabrera GP, Cruz Mendes A, Paraguai de Souza E,LYH, Trivellato FAA, Kawasaki JKA, et al. Effectiveotherapy against canine visceral leishmaniasis with the

    accine. Vaccine 2004;22(18):223443.Bulger PA, Dante S, Davis DvR, Perilli-Palmer B,

    lin RT. Characterization of canine humoral immuneses to outer surface protein subunit vaccines and to

  • Vaccines for leishmaniasis in the fore coming 25 years 1721

    natural infection by Lyme disease spirochetes. J Infect Dis1995;171:90915.

    [68] Chabalgoity JA, Moreno M, Carol H, Dougan G, Hor-maeche CE. Salmonella typhimurium as a basis for a liveoral Echinococcus granulosus vaccine. Vaccine 2001;19:4609.

    [69] VercamFollowtreateELISA

    [70] FernaJM. LIgG1/asymp2003;8

    [71] Reis ARC, MhallmBrazilchaga

    [72] Day Mleishmtation

    [73] ChamcytokiVet Im

    [74] VouldoGentilchemovia thChem

    [75] Reis AvalholeucocboneLeishm

    [76] Pinelliberg Egenereacti

    [77] BourdStollelymphinfect1997;5

    [78] MorenF, etviscerImmu

    [79] GuargMA, Ainfectciated2000;6

    [80] GuargCastilthe trImmu

    [81] Pinelliberg Egenereacti

    [82] LageCorreaspleengasi. V

    [83] AtletMappi

    identication of mutations in leishmaniasis-susceptible dogs.Infect Immun 2002;70(6):276371.

    [84] Quinnell RJ, Kennedy LJ, Barnes A, Courtenay O, Dye C,Garcez LM, et al. Suscetibility to visceral leishmaniasis indomestic dog is associated with MHC class II polymorphism.

    munessoaa leisev Bintun, et autaneelezntunensisolomrmijos W.utoclant a004;2hari, Mole dnthro998;3ome, Gharial outanehalilusa BentioCG-catti IE, etia maaccine Lucerthoeniciaccin999;1erreietectn Amis of003;4itcheLeis

    ressoishmayrinO, Guimiomericoheboubleatedgainsrict,enarilva Jn theiasisswalastesorrillmen F, Fernandez-Perez FJ, del Amo C, Alunda JM.-up of Leishmania infantum naturally infected dogsd with allopurinol: immunouorescence antibody test,and Western blot. Acta Trop 2002;84:17581.ndez-Perez FJ, Gomez-Munoz MT, Mendez S, Alundaeishmania-specic lymphoproliferative responses andIgG2 immunodetection patterns by Western blot intomatic, symptomatic and treated dogs. Acta Trop6:8391.B, Teixeira-Carvalho A, Vale AM, Marques MJ, Giunchettiayrink W, et al. Isotype patterns of immunoglobulins:arks for clinical status and tissue parasite density inian dogs naturally infected by Leishmania (Leishmania)si. Vet Immunol Immunopathol 2006;112:10216.J. Immunoglobulin G subclass distribution in canineaniosis: A review and analysis of pitfalls in interpre-. Vet Parasitol 2007;147:28.izo C, Moreno J, Alvar J. Semi-quantitative analysis ofne expression in asymptomatic canine leishmaniasis.munol Immunopathol 2005;103:6775.ukis I, Drapier JC, Nussler AK, Tselentis Y, Da Silva OA,ini M, et al. Canine visceral leishmaniasis: successfultherapy induces macrophage antileishmanial activitye L-arginine nitric oxide pathway. Antimicrob Agentsother 1996;40:2536.B, Teixeira-Carvalho A, Giuchetti RC, Guerra LL, Car-MG, Mayrink W, et al. Phenotypic features of circulatingytes as immunological markers for clinical status andmarrow parasite density in dogs naturally infected byania chagasi. Clin Exp Immunol 2006;146:30311.E, Van der Kaaij SY, Stappendel R, Fragio C, Ruiten-J, Bernardina W, et al. Detection of canine cytokineexpression by reverse transcription-polymerase chainon. Vet Immunol Immunopathol 1999;69:1216.oiseau G, Bonnefont C, Hoareau E, Boehringer C,T, Chabanne L. Specic IgG1 and IgG2 antibody andocyte subset levels in naturally Leishmania infantum-ed treated and untreated dogs. Vet Immunopathol9:2130.o J, Nieto J, Chamizo C, Gonzalez F, Blanco F, Barkeral. The immune response and PBMC subsets in canineal leishmaniasis before and after chemoterapy. Vetnopathol 1999;30:18195.a JL, Moreno J, Lucientes J, Gracia MJ, Peribanezlvar J, et al. Canine leishmaniasis transmission: higherivity among naturally infected dogs to sandies is asso-with lower proportions of T helper cells. Res Vet Sci9:24953.a JL, Moreno J, Lucientes J, Gracia MJ, Peribanez MA,lo JA. Evaluation of a specic immunochemotherapy foreatment of canine visceral leishmaniasis. Vet Immunolnopathol 2002;88:1320.E, van der Kaaij SY, Slappendel R, Fragio C, Ruiten-J, Bernadina W, et al. Detection of canine cytokineexpression by reverse transcription-polymerase chainon. Vet Immunol Immunopathol 1999;69:1216.RS, Oliveira GC, Busek SU, Guerra LL, Giunchetti RC,-Oliveira R, et al. Analysis of the cytokine prole incells from dogs naturally infected by Leishmania cha-et Immunol Immunopathol 2007;115:13545.L, Francino O, Solano-Gallego L, Renier C, Sanchez A.ng and sequencing of the canine NRAMP1 gene and

    Im[85] P

    nR

    [86] AMc

    [87] VAnC

    [88] Aloav2

    [89] SAga1

    [90] MFtc

    [91] KMvB

    [92] SEnV

    [93] DBgv1

    [94] FDas2

    [95] Maple

    [96] MAqa

    [97] MDtat

    [98] GSonO

    [99] CZogenetics 2003;55:238.SB, Pestana BR. Ensaio sobre a vacinacao preventivahmaniose tegumentar americana, com germes mortos.ol Hyg 1940;10:1128.es CM, Mayrink W, Magalhaes PA, Costa A, Melo MN, Diasl. Controlled eld trials of a vaccine against New Worldous leishmaniasis. Int J Epidemiol 1986;15:57280.ID, Gilchrist K, Arbelaez MP, Rojas CA, Puerta JA,es CM, et al. Failure of a killed Leishmania amazo-vaccine against American cutaneous leishmaniasis inbia. Trans R Soc Trop Med Hyg 2005;99:5938.s RX, Weigel MM, Calvopina M, Hidalgo A, Ceval-Correa J.Safety, immunogenecity, and efcacy of anaved Leishmania amazonensis vaccine plus BCG adju-gainst New World cutaneous leishmaniasis. Vaccine2:13206.I, FeKri AR, Aatonian MR, Khamesipour A, Nadim

    usavi MR, et al. Randomised vaccine trial of sin-ose of killed Leishmania major plus BCG againstponotic cutaneous leishmaniasis in Bam, Iran. Lancet51(9115):15403.ni AZ, Jalayer T, Emamjomeh M, Khamesipour A, Zickerssemi RL, et al. A randomised, double-blind, controlledf a killed L. major vaccine plus BCG against zoonoticous leishmaniasis in Iran. Vaccine 1999;17:46672.EAG, El Hassan AM, Zijlstra EE, Mukhtar MM, Ghalib HW,, et al. Autoclaved Leishmania major vaccine for pre-n of viscral leishmaniasis: a randomised, doubled-blind,ontrolled trial in Sudan. Lancet 2000;356:15659.N, Osman HY, Daifalla NS, Younis SA, Khalil EA, Zijlstraal. Immunogenicity and safety of autoclaved Leishma-jor plus BCG vaccine in healthy Sudanese volunteers.e 2001 Feb 28;19:21006.a PM, Mayrink W, Alves CR, Coutinho SG, Oliveira MP,AL, et al. Evaluation of the stability and immuno-

    ty of autoclaved and nonautoclaved preparations of ae against American tegumentary leishmaniasis. Vaccine7:117985.ra WA, Mayrink W, dos Mares-Guia ML, Tavares CA.ion and characterization of leishmania antigens fromerican cutaneous leishmaniasis vaccine for diagno-visceral leishmaniasis. Diagn Microbiol Infect Dis

    5(1):3543.ll GF, Handman E. The glycoconjugate derived fromhmania major receptor for macrophages is a sup-genic, disease-promoting antigen in murine cutaneousaniasis. Parasite Immunol 1986;8:25563.k W, Botelho ACC, Magalhaes PA, Batista SM, Limaenaro O, et al. Imunoterapia, imunoquimioterapia eterapia no tratamento da leishmaniose tegumentarana. Rev Soc Bras Med Trop 2006;39:1421.ali M, Khamesipour A, Mobedi I, Zarei Z, Fesharki RH.-blind randomized efcacy eld trial of alum precipi-autoclaved Leishmania major vaccine mixed with BCGt canine visceral leishmaniasis in Meshkin-Shahr dis-I.R. Iran. Vaccine 2004;22:4097100.o O, Pinto JA, Da Costa CA, Franca-Silva JC, Costa RT,C, et al. Phase III randomized double blind clinical trialefcacy of a vaccine agaisnt canine visceral leishma-in urban area of Montes Claros, MG, Brazil. Mem Instdo Cruz 1996;91:116.M, Blackwell J, Trujillo D, Formica S, Cabrera M,

    a G, et al. Immune response in healthy volunteers

  • 1722 C.B. Palatnik-de-Sousa

    vaccinated with killed leishmanial promastigotes plus BCG.I: Skin-test reactivity, T-cell proliferation and interferon-gamma production. Vaccine 1994;12(11):104151.

    [100] Convit J, Castellanos PL, Rondon A, Pinardi ME, Ulrich M,Castes M, et al. Immunotherapy versus chemotherapy inlocalis

    [101] CruzreplacS A 19

    [102] Souzafor thcana.

    [103] Alexancysteiulence1998;1

    [104] Papadlion I,biopteuated

    [105] MuyomM, pachallenant1998;1

    [106] DavouMastestrainlive ch

    [107] RequeRecen2004;4

    [108] Titus Rof artserveParasi

    [109] Xu D,tectiosuscepuated1995;8

    [110] ConneDG. Esis witLeishm1993;1

    [111] StreitLCR1suscep

    [112] RamrH, Somutan11 elic2001;2

    [113] McMahK, Bering GPImmun

    [114] Gonzajasvaaregimeing thBALB/15;20(

    [115] DunanQuilicleishmmania

    [116] Borja-Cabrera GP. Analise do potencial diagnostico,prognostico e imunoprotetor do antgeno FML (Ligante deFucose Manose) de Leishmania (L.) donovani, no calazarcanino experimental e de area endemica. PhD thesis. Uni-versidade Federal Fluminense. pp. 8198; 2000.a Silve Sofcacndemine 2orja-ouzagainsan e

    ine 2emesin G,eishmxcretastigemesugneaninendemeld talatn, Palntibond pryg 19orjaayrinnd pr999;6arrae-Souaccinine 2antosalatnant ieishmalatn, Borects hJ BraantosalatnurinaccinumonourthgainsrammDR/Palatn, Tinourin

    aja saliveiico Duillagains0.araivabreLeishissioed cutaneous leishmaniasis. Lancet 1987;1:4015.A, Coburn CM, Beverley SM. Double targeted geneement for creating null mutants. Proc Natl Acad Sci U91;88:71704.AE, Barres PA, Coombs GH, Mottram JC. Null mutantse lmcpa cysteine proteinase gene in Leishmania mexi-Mol Biochem Parasitol 1994;63:21320.der J, Coombs GH, Mottram JC. Leishmania mexicanane proteinase-decient mutants have attenuated vir-for mice and potentiate a TH1 response. J Immunol61:6794801.opoulou B, Roy G, Breton M, Kunding C, Dumas C, Fil-et al. Reduced infectivity of a Leishmania donovanirin transporter genetic mutant and its use as an atten-strain for vaccination. Infect Immun 2002;70:628.bwe A, Olivier M, Harvie P, Bergeron MG, Ouellettepadopoulou B. Protection against Leihsmania majornge infection in mice vaccinated with live recombi-parasites expressing a cytotoxic gene. J Infect Dis77:18895.di N, Tate CA, Warburton C, Murray A, mahboudi F, Mcr WR. Development of a recombinant Leishmania majorsensitive to ganciclovir and 5-uoro cytosine for use aallenge in clinical trials. Vaccine 2005;23:11707.na JM, Iborra S, Carrion J, Alonso C, Soto M.t advances in vaccines dfor leishmanasis. Exp Opin:150517.G, Bishop JV, Mejia JS. The immunomodulatory factorshropod saliva and the potential for theses factors toas vaccine targets to prevent pathogen transmission.te Immunol 2006;28:13141.McSorley SJ, Chateld SN, Dougan G, Liew FY. Pro-n against Leishmania major infection in geneticallytible BALB/c mice by gp63 delivered orally in atten-Salmonella typhimurium (AroA- AroD-). Immunology5:17.ll ND, Medina-Acosta E, McMaster WR, Bloom BR, Russellffective immunization against cutaneous leishmania-h recombinant bacille Calmette-Guerin expressing theania surface proteinase gp63. Proc Natl Acad Sci U S A5:114737.JA, Recker TJ, Donelson JE, Wilson ME. BCG expressingof Leishmania chagasi induces protective immunity intible mice. Exp Parasitol 2000;94:3341.ez JR, Gilchrist K, Robledo S, Sepulveda JC, Mollldati D, et al. Attenuated Toxoplasma gondii ts-4ts engineered to express the Leishmania antigen KMP-it a specic immune response in BALB/c mice. Vaccine0:45561.on-Pratt D, Rodriguez D, Rodriguez JR, Zhang Y, Mansongman C, et al. Recombinant vaccinia viruses express--46/M-2 protect against Leishmania infection. Infect1993;61:33519.

    lo RM, del Real G, Rodriguez JR, Rodriguez D, Hel-ra R, Lucas P, et al. A heterologous prime-boostusing DNA and recombinant vaccinia virus express-

    e Leishmania infantum P36/LACK antigen protectsc mice from cutaneous leishmaniasis. Vaccine 2002 Jan78):122631.S, Frommel D, Monjour L, Ogunkolade BW, Cruz A,

    i M, The Phocean veterinary study group on visceralaniasis. Vaccination trial against canine visceral leish-sis. Parasite Immunol 1989;11:397402.

    [117] ddEec

    [118] BSainc

    [119] Ltlem

    [120] LHce

    [121] PKaaH

    [122] BMa1

    [123] Pdvc

    [124] SPvl

    [125] PEt(

    [126] SPmv

    [127] DFagT

    [128] PEml

    [129] ONQa2

    [130] SC(ma VO, Borja-Cabrera GP, Correia Pontes NN, Paraguaiuza E, Luz KG, Palatnik M, et al. A Phase III trial ofy of the FML-vaccine against canine kala-azar in anic area of Brazil (Sao Goncalo do Amarante, RN). Vac-001;19:106881.Cabrera GP, Correia Pontes NN, da Silva VO, Paraguai deE, Santos WR, Gomes EM, et al. Long lasting protectiont canine kala-azar using the FML-QuilA saponin vaccinendemic area of Brazil (Sao Goncalo do Amarante). Vac-002;20:327784.re JL, Holzmuller P, Cavaleyra M, Goncalves RB, Hot-Papierok G. Prottection against experimental visceralaniasis infection in dogs immunized with purieded secreted antigens of Leishmania infantum pro-otes. Vaccine 2005;23:282540.re JL, Holzmuller P, Goncalves RB, Bourdoiseau G,t C, Cavaleyra M, et al. Long-lasting protection againstvisceral leishmaniasis using the LiESAp-MDP vaccine inic areas of France: double-blind randomised efcacyrial. Vaccine 2007;25:422334.ik de Sousa CB, Gomes EM, Paraguai de Souza E, Luzatnik M, Borojevic R. Leishmania donovani: titration ofdies to the fucose mannose ligand as an aid in diagnosisognosis of visceral leishmaniasis. Trans R Soc Trop Med95;89:3903.Cabrera GP, da Silva VO, da Costa RT, Barbosa Reis A,k W, Genaro O, et al. The FML-ELISA assay in diagnosisognosis of canine visceral leishmaniasis. Am J Trop Med1(2):296301.LE, Borja-Cabrera GP, Santos FN, Souza LOP, Palatnik-sa CB, Menz I. Safety trial using the Leishmune

    e against canine visceral leishmaniasis in Brazil. Vac-007;25:21806.WR, de Lima VMF, Paraguai de Souza E, Bernardo RR,

    ik M, Palatnik de Sousa CB. Saponins, IL12 and BCG adju-n the FML-vaccine formulation against murine visceralaniasis. Vaccine 2002;21:3043.ik de Sousa CB, Bunn Moreno MM, Paraguai de Souzaojevic R. The FML vaccine (fucose-manosse ligand) pro-amsters from experimental kala-azar. Ciencia e Culturaz Assoc Adv Sci) 1994;46:2906.WR, Aguiar IA, Paraguai de Souza E, de Lima VFM,

    ik M, Palatnik-de-Sousa CB. Immunotherapy againste experimental visceral leishmaniasis with the FML-e. Vaccine 2003;21(32):466876.teil E, McMahon-Pratt D, Price VL. Report of theTDR/IDRI Meeting on Second Generation vaccines

    t Leishmaniaisis. UNDP/EWorld Bank/WHO Special Pro-e for Research & Training in Tropical Diseases (TDR).RD/Leish/Vac/01.1.ik de Sousa CB, Santos WR, Casas CP, Paraguai de Souzaco LW, da Silva BP, et al. Protective vaccination against

    e visceral leishmaniasis using aldehyde-containing Quil-ponaria sapogenins. Vaccine 2004;22(19):24709.ra-Freitas E, Casas CP, Borja-Cabrera GP, Santos FN,, Souza LOP, et al. Acylated and deacylated saponins ofja saponaria mixture as adjuvants for the FML-vaccinet visceral leishmaniasis. Vaccine 2006;24:3909

    a EM, Mendes-Aguiar CO, Paraguai de Souza E, Borja-ra GP, Fampa P, Parra LE, et al. The FML-vaccinemune) against canine visceral leishmaniasis: a trans-n blocking vaccine. Vaccine 2006;24:242331.

  • Vaccines for leishmaniasis in the fore coming 25 years 1723

    [131] Marciani DJ. Vaccine adjuvants: role and mechanisms ofaction in vaccine immunogenicity. Drug Discov Today 2003;8(20):93443.

    [132] Nico D, Souza LOP, de Almeida LN, Monteiro ACS, ScharfsteinJ Palatnik-de-Sousa CB. 2007. Kinins generated upon vacci-nation

    are paagainsing 25Nethe

    [133] Stagerstage-induce2000;1

    [134] SkeikyJA, Sctein tImmu

    [135] Gurunmentsparasi

    [136] PintoIntrana parInfect

    [137] SoliozG, Maagainscine 1

    [138] Masinaagainskeys u2003;1

    [139] RafatiLebbacinesproteclymph

    [140] SjolanVaccinLeishmbut d2077

    [141] WilsonAli KMstimulImmu

    [142] GhoshproteiwhichVaccin

    [143] CampSeikyinduceman p2001;6

    [144] MorenC, etnia prleishm

    [145] Poot Ttrambaseddoes ntum. V

    [146] MolanSoto Mgenic

    dogs experimentally infected with L. infantum. Vet ImmunolImmunopathol 2003;92:113.

    [147] Badaro R, Lobo I, Munos A, Netto EM, Modabber F, Campos-Neto A, et al. Immunotherapy for drug-refractory mucosalleishmaniasis. J Infect Dis 2006;194:11519.halibeveloermaoi:10ontenolerellisoonsiseishmia elofectoler11f, aiscermun

    rado, Graishmia inffectoormandmhe pr001;1keikyraneulti-

    ormuougnt al.ciencalatnani sompo993;5hmedompaates004;2ampoeed Sishmaniamunorraaccinucleogains8;22(afatiive coith DaccinhosheishmNA vaesponragak. Immein painfeelbyonovwith the QS21 saponin-containing Leishmune vaccinertially responsible for induction of protective immunityt visceral leishmaniasis. Vaccine Congress. Celebrat-years of publication. 911 December, Amsterdam, therlands.S, Smith DF, Kaye PM. Immunization with a recombinantregulated surface protein from Leishmania donovanis protection against visceral leishmaniasis. J Immunol65:706471.YA, Kennedy M, Kaufman D, Borges MM, Guderian

    holler JK, et al. LeIF: a recombinant Leishmania pro-hat induces an IL-12-mediated Th1 cytokine prole. Jnol 1998;161:61719.athan S, Prussin C, Sacks DL, Seder RA. Vaccine require-for sustained cellular immunity to an intracellular

    tic infection. Nat Med 1998;4:140915.EF, Pinheiro RO, Rayol A, Larraga V, Rossi-Bergmann B.asal vaccination against cutaneous leishmaniasis withticulated leishmanial antigen or DNA encoding LACK.Immun 2004;72:45217.N, Blum-Tirouvanziam U, Jacquet R, Rafati S, Corradinuel J, et al. The protective capacities of histone H1t experimental murine cutaneous leishmaniasis. Vac-999;18:8509.

    S, Gicheru M, Demotz SO, Fasel NJ. Protectiont cutaneous leishmaniasis in outbred vervet mon-sing a recombinant histone H1 antigen. J Infect Dis88:12507.S, Kariminia A, Seyde-Eslami S, Narimani M, Taheri T,tard M. Recombinant cysteine proteinases-based vac-against Leishmania major in BALB/c mice: the partialtion relies on interferon gamma producing CD8(+) Tocyte activation. Vaccine 2002;20:243947.der A, Baldwin TM, Curtis JM, Bengtsson KL, Handman E.ation with recombinant Parasite Surface Antigen 2 fromania major induces a Th1 type of immune response

    oes not protect against infection. Vaccine 1998;16:84.ME, Young BM, Andersen KP, Weinstock JV, Metwali A,

    , et al. A recombinant Leishmania chagasi antigen thatates cellular immune responses in infected mice. Infectn 1995;63:20629.A, Zhang WW, Matlashewski G. Immunization with A2n results in a mixed Th1/Th2 and a humoral responseprotects mice against Leishmania donovani infections.e 2001;20:5966.os-Neto A, Porrozzi R, Greeson K, Coler RN, Webb JR,YA, et al. Protection against cutaneous leishmaniasisd by recombinant antigens in murine and nonhu-rimate models of the human disease. Infect Immun9:41038.o J, Nieto J, Masina S, Canavate C, Cruz I, Chicharroal. Immunization with H1, HASPB1 and MML Leishma-oteins in a vaccine trial against experimental canineaniasis. Vaccine 2007;25:5290300., Spreeuwenberg K, Sanderson SJ, Schijns VECJ, Mot-JC, Coombs GH, et al. Vaccination with a preparationon recombinant cystein pepetidases and canine IL-12ot protect dogs from infection with Leishmania infan-accine 2006;24:24608.o I, Alonso MG, Miron C, Redondo E, Requena JM,, et al. A Leishmania infantum multi-component anti-protein mixed with live BCG confers protection to

    [148] Gdddc

    [149] CncLnIn

    [150] C1vIm

    [151] GRlenin

    [152] NHt2

    [153] SCmf

    [154] MeS

    [155] Pvc1

    [156] Acd2

    [157] CRlemIm

    [158] IbVna2

    [159] RtwV

    [160] GLDr

    [161] FJtin

    [162] MdH, Modabber F. Consultation meeting on thepment of therapeutic vaccines for post kala-azarl leishmaniasis. Kinetoplastid Biol Dis 2007;6:7.1186/1475-9292-6-7. http:/www.kinetoplastids.com/t/6/1/7.RN, Skeiky YA, Bernards K, Greeson K, Carter D, Cor-n CD, et al. Immunization with a polyprotein vaccineting of the T-Cell antigens thiol-specic antioxidant,ania major stress-inducible protein 1, and Leishma-ngation initiation factor protects against leishmaniasis.Immun 2002;70:421525.RN, Goto Y, Bogatzki L, Raman V, Reed SG. Leish-recombinant polyprotein vaccine that protects against

    al Leishmaniasis by elicitation of CD4+ T cells. Infect2007;75:464854.

    ni L, Foglia Manzillo V, Pagano A, Piantedosi D, De Lunamiccia M, et al. Failure of a multi-subunit recombinantanial vaccine (MML) to protect dogs from Leishma-antum infection and to prevent disease progression ined. Vaccine 2005;23:524551.ohammadi AH, Hochrein H, Curtis JM, Baldwin TM,an E. Paradoxical effects of IL-12 in leishmaniasis inesence and absence of vaccinating antigen. Vaccine9:404352.YA, Coler RN, Brannon M, Stromberg E, Greeson K,RT, et al. Protective efcacy of a tandemly linked,subunit recombinant leishmanial vaccine (Leish-111f)lated in MPL adjuvant. Vaccine 2002;20:3292303.eau E, Altare F, Wakil AE, Zheng S, Coppola T, Wang ZE,Expression cloning of a protective Leishmania antigen.e 1995;268:5636.ik de Sousa CB, Dutra HS, Borojevic R. Leishmania dono-urface glycoproteins GP36, is the major immunogennent of the Fucose Mannose Ligand (FML). Acta Tropica3:5972.SB, Bahloul C, Robbana C, Askri S, Dellagi K. A

    rative evaluation of different DNA vaccine candi-against experimental murine due to L. major. Vaccine2(1314):16319.s-Neto A, Webb JR, Greeson K, Coler RN, Skeiky YA,G. Vaccination with plasmid DNA encoding TSA/LmSTI1anial fusion proteins confers protection against Leish-major infection in susceptible BALB/c mice. Infect2002;70:282836.S, Soto M, Carrion J, Alonso C, Requena JM.

    ation with a plasmid DNA cocktail encoding thesomal histones of Leishmania confers protectiont murine cutaneous leishmaniosis. Vaccine 2004 Sep2930):386576.S, Salmanian AH, Taheri T, Vafa M, Fasel N. A protec-cktail vaccine against murine cutaneous leishmaniasisNA encoding cysteine proteinases of Leishmania major.e 2001;19:336975.A, Labrecque S, Matlashewski G. Protection againstania donovani infection by DNA vaccination: increasedccination efciency through inhibiting the cellular p53se. Vaccine 2001;19:316978.i K, Sufa I, Ferrua B, Rousseau D, Le Fichoux Y, Kubarunisation with DNA encoding Leishmania infantum pro-pLe22 decreases the frequency of parasitemic episodescted hamsters. Vaccine 2001;19:17019.PC, Yang J, Zhao W, Perez LE, Cheng J. Leishmania

    ani p36(LACK) DNA vaccine is highly immunogenic but

  • 1724 C.B. Palatnik-de-Sousa

    not protective against experimental visceral leishmaniasis.Infect Immun 2001;69:471925.

    [163] Sukumaran B, Tewary P, Saxena S, Madhubala R. Vaccinationwith DNA encoding ORFF antigen confers protective immu-nity in mice infected with Leishmania donovani. Vaccine2003;21:12929.

    [164] Souza LOP, Palatnik-de-Sousa CB. Cross-protective effciacyof the prophylactic VR1012-NH36 Leishmania donovani DNAvaccine agsisnt cutaneous leishmaniasis by leishmanai ama-zonensis. Abstracts of the Vaccine Congress: 25 years ofpublication. 911 December, Amsterdam, the Netherlands,vol. 1, p. 168.

    [165] Borja Cabrera GP, Santos FN, Miyashiro LM, Santos FB, Palatnikde Sousa CB. 2007. Nucleoside hydrolase DNA vaccine againstvisceral leishmaniasis. Vaccine Congress. Celebrating 25 yearsof publication. 911 December, the Netherlands, vol. 1, p.167.

    [166] Basu R, Bhaumik S, Basu JM, Naskar K, De T, Roy S. Kinetoplas-tid membrane protein-11 DNA vaccination induces completeprotection against both pentavalent antimonial-sensitive and-resistant strains of Leishmania donovani that correlates withinducible nitric oxide synthase activity and IL-4 generation:evidence for mixed Th1- and Th2-like responses in visceralleishmaniasis. J Immunol 2005;174:716071.

    [167] Gamboa-Leon R, Paraguai de Souza E, Borja Cabrera GP, San-tos FN, Miyashiro LM, Pinheiro RO, et al. Immunotherapyagainst visceral leishmaniasis with the nucleoside hydrolase-DNA vaccine of L. donovani. Vaccine 2006;24:486373.

    [168] Campbell K, Diao H, Ji J, Soong L. DNA immunization withthe gene encoding P4 nuclease of Leishmania amazonensisprotects mice against cutaneous Leishmaniasis. Infect Immun2003;71:62708.

    [169] Handman E, Noormohammadi AH, Curtis JM, Baldwin T,Sjolander A. Therapy of murine cutaneous leishmaniasis byDNA vaccination. Vaccine 2000;18:30117.

    [170] Lima HC, Titus RG. Effects of sand y ve