KedzierskiHV7-11

download KedzierskiHV7-11

of 11

Transcript of KedzierskiHV7-11

  • 8/12/2019 KedzierskiHV7-11

    1/11

  • 8/12/2019 KedzierskiHV7-11

    2/11

    www.landesbioscience.com Human Vaccines 1205

    SPECIAL FOCUS REVIEW: NEGLECTED VACCINESDEVELOPING WORLD SPECIAL FOCUS REVIEW: NEGLEC TED VACCINESDEVELOPING WORLD

    complex, it is believed that human VL trials will follow any suc-cessful CL immunization program. Whether the same vaccinewill work against both forms of the disease is not clear at present.Literature reporting cross-protection between VL and CL speciesis scarce. Exposure to heat-killed L. donovanihas been shown toprotect against L. majorchallenge,14and recently immunizationwith the polyprotein KSAC with MPL-SE A as an adjuvant has

    been reported to be effective against L. infantumand L. major.15The genomic analysis of three Leishmania species, which causedistinct disease pathologies, showed that L. major, L. brazilien-sisand L. infantumgenomes are highly conserved and have veryfew species-specific genes.16However, there is a high degree ofvariability in the cross-protective immunity induced by infec-tion with different Leishmania species17,18 and VL specific vac-cines may provide a more successful intervention. To compoundthe issue further is the fact that despite causing cutaneous dis-ease, the old and new world parasites, L. majorand L. mexicana/L. amazonensis, respectively, are markedly different.19There aredifferences in virulence factors between these species,20,21and insurvival capacity in the presence of Th1 responses.22These find-

    ings highlight an interesting and poorly understood aspect ofparasite immunobiology, and may have connotations for the vac-cine development process for the old and new world leishmani-asis. Yet another challenge for the vaccine is to obtain protectionagainst VL even if it is efficacious against the different forms ofCL. Nevertheless, the mortality associated with VL suggests thatpriority should be given to anti-VL vaccine, but VL vaccinationstudies are hampered by the lack of a suitable animal model ofdisease.

    Initially, vaccines composed of whole killed parasites havebeen proposed as both prophylactic and therapeutic vaccines. Thetherapeutic application may be particularly important in cases of

    drug resistant disease. In general, the whole-cell, killed vaccineshave been rather poorly defined and variable in potency, hencethey have rendered inconclusive results.12In addition, it has beendemonstrated that inoculation of killed parasites into immunemice leads to a loss of infection induced immunity.23This situa-tion might be analogous to that observed in endemic areas, wheremany individuals with subclinical leishmaniasis were vaccinatedwith killed vaccines, which subsequently led to a loss of natu-rally acquired immunity and vaccination failure. Although thishypothesis has not been proven, it clearly demonstrates that thereare still many unknown aspects of the vaccine design that wouldneed to be resolved prior its use in clinics. Nevertheless, the tri-als completed so far demonstrated their good safety profile, and

    despite poor prophylactic outcomes, showed encouraging resultsas therapeutic vaccines in South America and Sudan.

    A variety of different molecules (summarized in Table 1) hasbeen tested as second-generation vaccines,13,24and these includedantigens such as surface expressed glycoprotein leishmaniolysin(gp63) delivered by a plethora of immunization regimens, how-ever, promising findings from animal models were overshadowedby mostly negative T-cell responses in humans.25Nevertheless,this molecule is still being considered a strong candidate for avaccine and recent studies demonstrated its protective efficacy ina mouse model in combination with Hsp70 26and in heterologous

    on the observation that following lesion healing, an individual isresistant to reinfection. This method involved deliberate infec-tion of an individual using live parasites and has now been largelydiscontinued based on the grounds of quality control, parasitepersistence, emergence of HIV and ethical reasons, among theothers. The first-generation vaccines based on killed parasiteshave replaced leishmanization, but this type of vaccines have

    shown poor efficacy in clinical trials and in general the tradi-tional vaccine approaches have worked poorly.12The focus is nowon the second generation vaccines including genetically modi-fied parasites, defined subunit vaccines or recombinant bacteriaand viruses expressing leishmanial antigens.11 So far, their effi-cacy in the field trials has not been reported. Leishmania vaccinedevelopment has proven to be a difficult and challenging task,which is mostly hampered by inadequate knowledge of parasitepathogenesis and the complexity of immune responses needed forprotection. These aspects are of key importance in the vaccinedevelopment process. These issues as well as issues of fundingand knowledge dissemination in antileishmanial vaccine pro-grams have been highlighted in a recent proposal for a research

    agenda that stemmed from the International Symposium onLeishmaniasis Vaccines.13 In general, leishmaniasis is a diseaselinked to poverty, and is associated with malnutrition, poorhousing, illiteracy and lack of resources. Therefore, the targetpopulation for antileishmanial vaccine cannot afford the costof treatment, which surpasses a substantial percentage of house-hold income and would exert a significant financial burden onpeople in need for antileishmanial treatment. Hence, develop-ment of such a vaccine is considered unattractive to the industrydue to poor return of funds invested in research and develop-ment. Currently, leishmaniasis is considered to be endemic in 88countries, with 90% of VL cases occurring in Bangladesh, Brazil,

    India, Nepal and Sudan, and 90% CL cases in Afghanistan,Brazil, Iran, Peru, Saudi Arabia and Syria. Therefore, popula-tions of these countries might be the best choices for implemen-tation of new vaccination programs since the burden of disease inthose countries is quite significant. In several foci of the diseasein Africa, the prevalence of infection can reach up to 5% and notreatment is available, therefore a successful vaccine would be thebest method of control. However, unlike African countries, Indiaand Brazil have developed strong vaccine industries in order toachieve self-sufficiency in vaccine supply, which might be benefi-cial to anti-leishmanial vaccine program implementation.

    Overview of Vaccine Development

    Leishmaniasis is a disease that is most likely to be controlled by asuccessful vaccination program. Evidence from studies in animalmodels, indicates that protection can be achieved upon immu-nization with various vaccine formulations, however, to datesuch vaccines have been disappointing when tested in the field.11Historically, CL has been the focus of vaccination attempts, asit has been known for centuries that people who resolve a pri-mary CL skin lesion are protected from further infections.Vaccination against VL has received limited attention comparedwith CL, and although the demands for a VL vaccine are more

  • 8/12/2019 KedzierskiHV7-11

    3/11

    1206 Human Vaccines Volume 7 Issue 11

    Table 1.Summary of vaccination approaches discussed in the text

    Antigen Vaccine formulationVaccinated

    species

    Outcome of

    vaccinationTargeted disease Reference

    KSACpolyprotein (KMP11/SMT/A2/CPB) + monophosphoryl

    lipid Amouse protection VL/CL 15

    gp63 recombinant protein mouse no protection CL 132

    recombinant proteinnon-human

    primatepartial protection CL 133

    native protein mouse protection CL 134

    protein expressed in BCG mouse protection CL 135

    protein expressed in Salmonella mouse protection CL 136

    DNA vaccine mouse protection CL 137

    DNA vaccine mouse partial protection CL 138

    DC pulsed with native protein mouse protection CL 139

    recombinant protein/DNA prime/boost mouse protection VL 27

    HSP70 native proteins mouse protection VL 26

    gp46/PSA-2 native protein mouse protection CL 30,32

    recombinant protein mouse no protection CL 31

    DNA vaccine mouse no protection CL 140

    DNA vaccine mouse protection CL 32

    DNA vaccine mouse partial protection CL 138

    recombinant protein + IL-12 mouse protection CL 34

    p36/LACK DNA vaccine + protein expressed in vaccinia virus mouse protection CL 141

    protein expressed in Listeria monocytogenes mouse partial protection CL 142

    DNA vaccine mouse no protection VL 38

    DNA vaccine + protein expressed in vaccinia virus mouse protection VL 143

    DNA vaccine + protein expressed in vaccinia virus dog protection CVL 144

    DNA vaccine mouse no protection CL 138

    DNA vaccine + protein expressed in vaccinia virus mouse protection CL 145

    DNA vaccine + protein expressed in Salmonella mouse protection CL 146

    DNA vaccine mouse protection CL 147

    DNA vaccine mouse no protection CL 140

    DC pulsed with native antigen mouse protection CL 139

    recombinant protein fused with HIV TAT delivered in

    pulsed DCmouse protection CL 37

    CP recombinant protein mouse partial protection CL 148

    A2, P4, P8 native antigen mouse partial protection CL 40

    KMP-11 DNA vaccine hamster protection VL 41

    DC pulsed with native antigen mouse protection CL 139

    LCR1 protein expressed in BCG mouse partial protection VL 42

    HASPB1 recombinant protein mouse protection VL 43

    ORFF recombinant protein mouse partial protection VL 44

    DNA vaccine + recombinant protein mouse protection VL 44

    DNA vaccine mouse protection VL 149

    P0 DNA vaccine mouse protection CL 45

    PFR-2 DNA vaccine mouse protection CL 46

    NH36 DNA vaccine mouse partial protection VL/CL 47

    PPG recombinant protein hamster protection VL 48

    CL, cutaneous leishmaniasis; VL, visceral leishmaniasis; ML, mucosal leishmaniasis; CVL, canine visceral leishmaniasis; DC, dendritic cells; BCG, Mycobac-

    teriumbovisBacillus Calmette-Guerin.

  • 8/12/2019 KedzierskiHV7-11

    4/11

    www.landesbioscience.com Human Vaccines 1207

    response toward detrimental Th2 responses,35however, suscep-tible BALB/c mice immunised with LACK had the ability tocontrol a subsequent infection with L. major.36Recently, a fusionof LACK with HIV-1 TAT transduction domain delivered indendritic cells has been shown to be superior to immunization

    with LACK alone and improved disease outcome.37

    Nevertheless,the protective efficacy of LACK has been mainly demonstrated inthe L. majormodel, and LACK failed to protect against visceralleishmaniasis.38

    Other antigens tested against either CL or VL include amasti-gote cysteine proteases (CP),39cysteine proteinase A2 and amas-tigote membrane proteins P4 and P8,40kinetoplastid membraneprotein-11 (KMP-11),41amastigote LCR1,42hydrophilic acylatedsurface protein B1 (HASPB1),43 leishmanial antigen ORFF,44acidic ribosomal protein P0,45paraflagellar rod protein 2 (PFR-2),46NH36, a main component of the fucose-mannose ligand,47

    prime-boost approach using DNA and recombinant protein.27Another vaccine candidate has been a GPI-anchored membraneprotein gp46 or Parasite Surface Antigen 2 (PSA-2), that belongsto a gene family present in all Leishmania species except L. bra-ziliensis.28PSA-2 is involved in macrophage invasion through the

    interaction of its leucine rich repeats with complement receptor3.29 Immunization with the native polypeptides derived frompromastigotes protected mice against infection,30 but vaccina-tion with a recombinant protein derived from either promasti-gotes or amastigotes protein showed lack of protective efficacy.31Similarly, DNA vaccination conferred protection in mice whenused as either prophylactic32 or therapeutic vaccines.33 Anotherextensively tested antigen is the Leishmania homolog for recep-tors of activated C kinase (LACK) that is expressed through-out leishmanial life cycle.34 Immunization with LACK appearsto promote the expansion of IL-4 secreting T cells skewing the

    Table 1.Summary of vaccination approaches discussed in the text

    Antigen Vaccine formulationVaccinated

    species

    Outcome of

    vaccinationTargeted disease Reference

    ATP synthase

    chain, -tubulin,

    HSP70-related

    protein 1

    native protein in liposomes mice protection VL 49

    -glutamylcysteinesynthetase recombinant protein mouse protection VL 50

    P1 DNA vaccine, recombinant protein hamster partial protection VL 51

    Leish-111f recombinant polyprotein mouse protection CL 53

    recombinant polyprotein mouse protection VL 54

    recombinant polyprotein dog no protection CVL 55

    recombinant polyprotein + Glucantime dog protection CVL 56

    Leish-110f recombinant polyprotein mouse protection CL/VL 57

    Leish-F1 recombinant polyproteinhuman

    Phase I

    safe and immuno-

    genicCL 150

    recombinant polyprotein + sodium stibogluconatehuman

    Phase I

    safe and immuno-

    genicML 60

    recombinant polyprotein + meglumine antimoniate humanPhase I

    safe and immuno-genic

    CL 61

    recombinant polyproteinhuman

    Phase I

    safe and immuno-

    genicVL 59

    maxadilan synthetic protein mouse protection CL 64

    SP15 native protein/DNA vaccine mouse protection CL 65

    LJM19 DNA vaccine hamster protection VL 66

    dhfr-ts/ live attenuated parasites mouse protection CL 72

    mouse no protection CL 73

    cpa/ live attenuated parasites mouse protection CL 74

    hamster protection CL 75

    lpg2/ live attenuated parasites mouse protection CL 76

    LdCen1/ live attenuated parasites mouse protection VL 81

    PMM live attenuated parasites mouse protection CL 82

    SIR2+/ live attenuated parasites mouse protection VL 83

    CL, cutaneous leishmaniasis; VL, visceral leishmaniasis; ML, mucosal leishmaniasis; CVL, canine visceral leishmaniasis; DC, dendritic cells; BCG, Mycobac-

    teriumbovisBacillus Calmette-Guerin.

    (continued)

  • 8/12/2019 KedzierskiHV7-11

    5/11

    1208 Human Vaccines Volume 7 Issue 11

    an effective antileishmanial response might be linked to neutral-ization of saliva components.69However, recent insights into pro-tection induced by salivary gland components indicate that theprotective effect might be limited to a short-term exposure,70andthat the protective efficacy of saliva-based vaccine originating fromcolonized sand fly populations my be affected by variable responsesfollowing natural exposure.71

    In view of disappointing outcomes from field trials of wholecell killed vaccines and the general inability of subunit vaccinesto trigger long-term immunity, the live-attenuated vaccine pro-vides an appealing alternative, but only a few attenuated strainshave been tested so far. Avirulent microorganisms can be gen-erated by a defined genetic alteration, eliminating the risk ofparasite reversion to the virulent phenotype. Vaccination withdihydrofolate reductase thymidylate synthase (dhfr-ts) knockoutparasites led to protection in a mouse model,72but failed in theprimate model.73Deletion of cysteine proteinases in L. mexicanaled to an attenuated strain capable of triggering partial protec-tion against challenge in animal models.74,75 These moderatelyencouraging results were thought to be due to rapid elimination

    of parasites by the host, since knockout parasites were not persis-tent. Conversely, L. majorparasites lacking the lpg2gene persistedin mice without pathology and were able to confer protectionagainst infection,76 although required an adjuvant to be ableto protect resistant C57BL/6 mice.77However, over time thesemutants regained their ability to cause disease in the absence ofthe lpg2gene through an unknown compensatory mechanism,78suggesting that persistence may not be a desirable feature of alive-attenuated vaccine. It is already known that generation ofmemory cells during Leishmania infection does not require para-site persistence,79 therefore non-persistent attenuated mutantsmight offer a safer alternative. L. donovanicentrin null mutants

    (LdCEN

    -/-

    ) have been reported to have selective growth arrestin the amastigote stage of development, but were viable in cul-ture as promastigotes.80These mutants were unable to survive invitro in human macrophages and immunization with LdCen1-/-

    knockouts protected mice against L. donovaniand L. brazilien-sis infections.81 We have also demonstrated that non-persistentphosphomannomutase (PMM) mutants were able to protect sus-ceptible mice against L. majorchallenge by early suppression ofIL-10 and IL-13 production and increased magnitude of T-cellresponses.82These parasites are viable in vitro, but do not sur-vive in macrophages or in vivo in mice, similarly to LdCEN -/-

    parasites. The genetic attenuation does not necessarily require theproduction of null mutants. A single knockout of L. infantum

    SIR2gene was sufficient to prevent amastigotes from undergoingintracellular replication in macrophages. Immunization with themutant triggered strong T-cell responses and conferred completeprotection in a VL mouse model.83A limitation of this approachis the presence of the second SIR2allele making reversion to viru-lence a likely occurrence. An interesting alternative to geneticallyattenuated strains is the use of non-pathogenic species such asL. tarentolaeas live vaccines, an approach that has been provensuccessful in mice against VL.84 The use of Leishamania non-pathogenic species might be equivalent to the role vaccinia virusplayed in eradication of smallpox.

    and proteophosphoglycan (PPG).48 In addition, molecules suchas ATP synthase chain, -tubulin and heat shock 70-relatedprotein 1 precursor have been identified as novel vaccine can-didates.49 More recently, gamma-glutamylcysteine synthetase50and ribosomal P1 gene51have been shown to protect mice againstL. donovani.

    To date, only one second generation vaccine, Leish-111f, has

    been assessed in clinical trials.52 Initial immunization experimentsin a mouse model demonstrated that Leish-111f was able to pro-tect mice against L. major and L. amazonensis infection.53 Thereis some evidence that the Leish-111f vaccine can also induce par-tial protection against VL in animal models,54however, Leish-111ffailed to protect dogs against infection and did not prevent diseasedevelopment in a recent Phase III trial in dogs,55but was effectiveas therapeutic vaccine in combination with chemotherapy.56 Anoptimized version of the original construct, Leish-110f, has demon-strated strong immunogenicity and some protective efficacy againstL. infantumin mice,57and has been tested in dogs as a therapeu-tic vaccine in combination with chemotherapy and led to reducednumber of deaths and higher survival probability.58The Leish-111f

    vaccine is moving forward into clinical trials as LeishF1, F2 and F3,and is being trialled in combination with the MPL-SE adjuvant. Arecent small scale clinical trial in a L. donovaniendemic area showedLeish-F1-MPL-SE was safe and well-tolerated in people regardlessof their prior VL exposure, and induced strong antigen-specificT-cell responses.59In addition, Leish-F1 has been shown to be safeand immunogenic when used in combination with pentavalentantimonials in CL and mucosal leishmaniasis patients.60,61HumanPhase I and II clinical trials of Leish-111f have been completed overthe past few years in Brazil, Peru, Columbia and India. New clinicaltrials of Leish-F2 are to be conducted against CL in Peru and VL inSudan (clinicaltrials.gov).

    The sand fly injects Leishmania parasites in the presence ofsaliva, which contains a range of pharmacologically active mole-cules that can modulate hosts immune and inflammatory responsesand facilitate establishment of infection. For a number of years sali-vary gland antigens have been targeted as potential candidates forantileishmanial vaccine development, primarily against L. major.62Prior exposure of mice to bites of uninfected sand flies conferredprotection from L. major infection.63 Immunization with mol-ecules present in saliva, such as maxadilan64or a 15 kDa protein,SP15 65also induced protection against CL. It has been shown thatsalivary proteins, such as LJM19, protected hamsters from VL,66and immunization of dogs with salivary antigens led to the devel-opment of high IgG2 antibody levels and significant IFN pro-

    duction.67

    Peters et al.68

    demonstrated that sand fly transmissionof parasites abrogates vaccine-induced protective immunity. Whilemice vaccinated with killed parasites were refractory to a needlechallenge, they were susceptible to the sand fly inoculum imply-ing that the protective responses in vaccinated animals were eithernot generated or not maintained. These data provide a rationalefor the inclusion of sand fly saliva components, which are specificto natural infection, in vaccine design. In support of this notion,it has been shown that children who underwent anti-VL delayedtype hypersensitivity (DTH) conversion also had increased titersof antibodies directed to sand fly saliva, suggesting that mounting

  • 8/12/2019 KedzierskiHV7-11

    6/11

  • 8/12/2019 KedzierskiHV7-11

    7/11

    1210 Human Vaccines Volume 7 Issue 11

    re-activation of CD8+T cells is currently being developed againstVL (Paul Kaye, personal communication).

    A relat ively new vaccine platform that has been recentlytested against leishmaniasis utilizes an engineered nanopar-ticle delivery system. A number of nanoparticle-based thera-peutics are currently in use in the clinic, several of which arebased on liposomes, which have low immunogenicity, excellent

    safety profiles and established clinical-scale manufacturingprocess.124Such delivery system can also be used to carry anti-gens and make good vaccines.125Recently, various nanoparticlebased systems were used to deliver cysteine proteinases126 orsuperoxide dismutase B1 127and demonstrated feasibility of theapproach and exciting potential for developing new vaccinetechnology.

    An antileishmanial vaccine faces a challenging task of induc-ing protective immunity. As parasites persist following naturalinfection providing a low level of sustained antigenic stimula-tion, the immunity that is induced by Leishmania is analogousto concomitant immunity, which puts a considerable constrainton vaccine efficacy. T-cell subsets contributing to such immunity

    include CD4+T cells of the central memory79and effector Th1 128phenotypes, as well as CD8+T cells, which can play a critical rolein recall responses to secondary infection.129Therefore, an effica-cious vaccine would need to generate these subsets, but how togenerate these cells by vaccination remains a challenge. It is also atpresent unknown, which T cells may provide the best protectionand are most likely to survive long-term to fulfill their intendedrole. Recent studies suggest that the most protective CD4+T cellsare polyfunctional, capable of producing not just one, but severalcytokines.130CD4+ T cells of single specificity, secreting IFNonly, have limited capacity to provide durable protection againstLeishmania, whereas the ability of CD4+T cells to produce mul-

    tiple cytokines (IFN

    , TNF and IL-2) greatly increases the qual-ity of vaccine mediated responses.131In addition, elimination ofIL-10 promotes resistance, as this immunosuppressive cytokineappears to limit the generation of the protective T-cell subsetsfollowing vaccination.131Unfortunately, although there seem tobe clearly defined immunobiological goals for a vaccine, to datenone of the approaches have been successful in eliciting desirableprotective responses in humans.

    Concluding Remarks

    Preventive vaccines are recognized as the best and most cost-effective protection measure against pathogens, and are sav-

    ing millions of lives every year across the globe. Leishmaniavaccine development has proven to be a difficult and challeng-ing task, which has been hampered an inadequate knowledgeof disease pathogenesis, the complexity of immune responsesneeded for protection and the cost of vaccine development. Theburden of the disease is concentrated in resource poor nations,and a lack of political will and philanthropic investment fur-ther aggravate the situation. However, the rise of biotechnol-ogy industries in endemic countries, such as India and Brazil,may provide an impetus for a vaccine development. There arealso new European based vaccine efforts including a synthetic

    complexity of the parasite, the capacity to respond to several tar-gets may be a crucial prerequisite for a vaccine. Many candidateshave been selected based on the abundance, but this criteriondoes not necessarily correspond to their antigenicity. In termsof expression patterns, proteomic approaches provide valuableinformation for vaccine research, due to the importance of post-transcriptional regulation of gene expression. Improvements of

    proteomic analysis, incorporating the use of subcellular fractionsseems to allow the identification of novel stage specific proteins.For example, the proteome-serological approach, can providea useful tool to identify highly antigenic, but poorly expressedproteins and provide a catalog of antigenic patterns as well asimmune response specificity patterns that may advance develop-ment of vaccines.111Proteomic approach combined with reversevaccinology112is also being employed in a search of new vaccinecandidates and thus far has successfully been used to identify twonovel antigens.113T-cell epitope prediction is yet another power-ful approach, that might accelerate antigen discovery and furtherrefinement of prediction tools will improve their reliability,114,115however, at this stage experimental verification of the candidates

    is a necessity and a major limiting step considering a large num-ber of antigens that can be identified from such analyses.

    Vaccine formulations will be pivotal for vaccine efficacy.Several strategies have been tested, but in general the tradi-tional approaches have worked poorly. Virally vectored vac-cines emerged as a very effective and eliciting robust responsesplatforms that might address the deficiencies of traditionaldelivery systems, particularly where cell mediated responses areneeded for protection.116 Recently, our group utilized a modelof recombinant influenza expressing a single, immunodomi-nant LACK

    158173CD4 +T-cell peptide, and demonstrated that a

    prime/boost approach resulted in considerable protection against

    Leishmania in a mouse model and was associated with increasedIFNproduction by CD4+T cells (Kedzierska et al. manuscriptsubmitted). Influenza viruses are attractive candidates as vaccinevectors, with the approach being tried so far for HIV,117tubercu-losis,118malaria119and cancer.120These results point to the valueof recombinant influenza vector for Leishmania vaccination.Influenza viruses can be easily manipulated by a reverse genet-ics strategy121 to elicit prominent CD8+T-cell responses. Thus,the delivery of leishmanial epitopes in the context of influenzavector can facilitate the class I presentation in addition to classII presentation of a recombinant peptide. As the vector can bemanipulated by reverse genetics to include multiple epitopes, thisapproach has the capacity to generate broad protective responses

    with the potential to overcome HLA restriction and provideimmunological coverage to the broader host population. A poten-tial obstacle for a vaccine based on a viral vector is safety. In caseof on influenza there is already a precedent in using live, tem-perature sensitive influenza virus in a FluMist vaccine approvedby the FDA (live-attenuated, intranasal vaccine).122 Moreover,the insertion of foreign sequences into the neuraminidase (NA)segment results in a virus that can express the recombinant pep-tide as part of the NA protein, but cannot release virus progenyfrom infected cells, rendering the virus highly attenuated.123Anadenovirus-based therapeutic vaccine targeting the induction/

  • 8/12/2019 KedzierskiHV7-11

    8/11

    www.landesbioscience.com Human Vaccines 1211

    in research and development to move promising vaccine leadsalong the development pathway toward an effective and afford-able antileishmania vaccine.

    Acknowledgements

    I would like to thank Dr. Katherine Kedzierska for insightful andcritical review of the manuscript.

    vaccine RAPSODI (www.fp7-rapsodi.eu) and a DNA basedLEISHDNAVAX (www.leishdnavax.org). New adjuvants arealso being developed and there are several clinical vaccine tri-als in progress and in planning.5Given the rapid progress inthe fields of parasite immunology and genomics, a successfulanti-Leishmania vaccine should be achievable sooner ratherthan later, however, there is a clear need for greater investment

    References

    1. Handman E. Cell biology of Leishmania. Adv Parasitol1999; 44:1-39; PMID:10563394; DOI:10.1016/S0065-308X(08)60229-8.

    2. Chevalier B, Carmoi T, Sagui E, Carrette P, Petit D,De Mauleon P, et al. Report of the first cases of cutane-ous leishmaniasis in East Timor. Clin Infect Dis 2000;30:840; PMID:10816165; DOI:10.1086/313789.

    3. Kongkaew W, Siriar ayaporn P, Leelayoova S,Supparatpinyo K, Areechokchai D, Duang-ngern P, etal. Autochthonous visceral leishmaniasis: a report of asecond case in Thailand. Southeast Asian J Trop MedPublic Health 2007; 38:8-12; PMID:17539239.

    4. Rose K, Curtis J, Baldwin T, Mathis A, Kumar B,Sakthianandeswaren A, et al. Cutaneous leishmaniasisin red kangaroos: isolation and characterisation of the

    causative organisms. Int J Parasitol 2004; 34:655-64;PMID:15111087; DOI:10.1016/j.ijpara.2004.03.001.

    5. Modabber F. Leishmaniasis vaccines: past, presentand future. Int J Antimicrob Agents 2010; 36:58-61; PMID:20801000; DOI:10.1016/j.ijantimi-cag.2010.06.024.

    6. Dantas-Torres F. Canine leishmaniosis in SouthAmerica. Parasit Vectors 2009; 2:1; PMID:19426440;DOI:10.1186/1756-3305-2-S1-S1.

    7. Bern C, Maguire JH, Alvar J. Complexities of assess-ing the disease burden attributable to leishmaniasis.PLoS Negl Trop Dis 2008; 2:313; PMID:18958165;DOI:10.1371/journal.pntd.0000313.

    8. Patz JA, Graczyk TK, Geller N, Vittor AY. Effects ofenvironmental change on emerging parasitic diseases.Int J Parasitol 2000; 30:1395-405; PMID:11113264;DOI:10.1016/S0020-7519(00)00141-7.

    9. Pavli A, Maltezou HC. Leishmaniasis, an emerginginfection in travelers. Int J Infect Dis 2010; 14:1032-9;PMID:20952234; DOI:10.1016/j.ijid.2010.06.019.

    10. Kedzierski L, Sakthianandeswaren A, Curtis JM,Andrews PC, Junk PC, Kedzierska K. Leishmaniasis: cur-rent treatment and prospects for new drugs and vaccines.Curr Med Chem 2009; 16:599-614; PMID:19199925;DOI:10.2174/092986709787458489.

    11. Kedzierski L, Zhu Y, Handman E. Leishmania vac-cines: progress and problems. Parasitology 2006;133:87-112; PMID:17274851; DOI:10.1017/S0031182006001831.

    12. Noazin S, Modabber F, Khamesipour A, SmithPG, Moulton LH, Nasseri K, et al. First genera-tion leishmaniasis vaccines: a review of field efficacytrials. Vaccine 2008; 26:6759-67; PMID:18950671;DOI:10.1016/j.vaccine.2008.09.085.

    13. Costa CH, Peters NC, Maruyama SR, de Brito EC

    Jr, Santos IK. Vaccines for the leishmaniases: pro-posals for a research agenda. PLoS Negl Trop Dis2011; 5:943; PMID:21468307; DOI:10.1371/journal.pntd.0000943.

    14. Bebars MA, el Serougi AO, Makled KM, Mikhael EM,Abou Gamra MM, el Sherbiny M, et al. An experi-mental vaccine providing heterologous protection forLeishmania species in murine model. J Egypt SocParasitol 2000; 30:137-56; PMID:10786026.

    15. Goto Y, Bhatia A, Raman VS, Liang H, MohamathR, Picone AF, et al. KSAC: the first defined poly-protein vaccine candidate for visceral leishmani-asis. Clin Vaccine Immunol 2011; 18:1118-24;PMID:21632891; DOI:10.1128/CVI.05024-11.

    16. Peacock CS, Seeger K, Harris D, Murphy L, Ruiz JC,Quail MA, et al. Comparative genomic analysis ofthree Leishmania species that cause diverse human dis-ease. Nat Genet 2007; 39:839-47; PMID:17572675;DOI:10.1038/ng2053.

    17. Lainson R, Shaw JJ. Leishmaniasis in Brazil: XII.Observations on cross-immunity in monkeys and maninfected with Leishmania mexicana mexicana, L. m.amazonensis, L. braziliensisbraziliensis, L. b. guyanensisand L. b. panamensis. J Trop Med Hyg 1977; 80:29-35;PMID:405504.

    18. Porrozzi R, Teva A, Amaral VF, Santos da Costa MV,Grimaldi G Jr. Cross-immunity experiments betweendifferent species or strains of Leishmania in rhesusmacaques (Macaca mulatta). Am J Trop Med Hyg2004; 71:297-305; PMID:15381810.

    19. McMahon-Pratt D, Alexander J. Does the

    Leishmania major paradigm of pathogenesis and pro-tection hold for New World cutaneous leishmani-ases or the visceral disease? Immunol Rev 2004;201:206-24; PMID:15361243; DOI:10.1111/j.0105-2896.2004.00190.x.

    20. Ilg T. Lipophosphoglycan is not required for infec-tion of macrophages or mice by Leishmania mexi-cana.EMBO J 2000; 19:1953-62; PMID:10790362;DOI:10.1093/emboj/19.9.1953.

    21. Spth GF, Epstein L, Leader B, Singer SM, Avila HA,Turco SJ, et al. Lipophosphoglycan is a v irulence factordistinct from related glycoconjugates in the protozoanparasite Leishmania major. Proc Natl Acad Sci USA2000; 97:9258-63; PMID:10908670; DOI:10.1073/pnas.160257897.

    22. Soong L, Chang CH, Sun J, Longley BJ Jr, RuddleNH, Flavell RA, et al. Role of CD4+T cells in patho-genesis associated with Leishmania amazonensis infec-tion. J Immunol 1997; 158:5374-83; PMID:9164958.

    23. Okwor I, Liu D, Beverley SM, Uzonna JE. Inoculationof killed Leishmania major into immune mice rap-idly disrupts immunity to a secondary challenge viaIL-10-mediated process. Proc Natl Acad Sci USA2009; 106:13951-6; PMID:19666482; DOI:10.1073/pnas.0905184106.

    24. Nagill R, Kaur S. Vaccine candidates for leish-maniasis: A review. Int Immunopharmacol 2011;11:1464-88; PMID:21616175; DOI:10.1016/j.intimp.2011.05.008.

    25. Russo DM, Burns JM Jr, Carvalho EM, ArmitageRJ, Grabstein KH, Button LL, et al. Human T cellresponses to gp63, a surface antigen of Leishmania. JImmunol 1991; 147:3575-80; PMID:1940356.

    26. Kaur T, Sobti RC, Kaur S. Cocktail of gp63 andHsp70 induces protection against Leishmania don-

    ovani in BALB/c mice. Parasite Immunol 2011;33:95-103; PMID:21226722; DOI:10.1111/j.1365-3024.2010.01253.x.

    27. Mazumder S, Maji M, Das A, Ali N. Potency, effi-cacy and durability of DNA/DNA, DNA/protein andprotein/protein based vaccination using gp63 againstLeishmania donovani in BALB/c mice. PLoS ONE2011; 6:14644; PMID:21311597; DOI:10.1371/jour-nal.pone.0014644.

    28. McMahon-Pratt D, Traub-Cseko Y, Lohman KL,Rogers DD, Beverley SM. Loss of the GP46/M-2 surfacemembrane glycoprotein gene family in the Leishmaniabraziliensis complex. Mol Biochem Parasitol 1992;50:151-60; PMID:1542309; DOI:10.1016/0166-6851(92)90252-F.

    29. Kedzierski L, Montgomery J, Bullen D, Curtis J,Gardiner E, Jimenez-Ruiz A, et al. A leucine-rich repeatmotif of Leishmania parasite surface antigen 2 bindsto macrophages through the complement receptor 3. JImmunol 2004; 172:4902-6; PMID:15067069.

    30. Handman E, Symons FM, Baldwin TM, Curtis JM,Scheerlinck JP. Protective vaccination with promasti-gote surface antigen 2 from Leishmania majoris medi-ated by a TH1 type of immune response. Infect Immun1995; 63:4261-7; PMID:7591056.

    31. Sjlander A, Baldwin TM, Curtis JM, Bengtsson KL,Handman E. Vaccination with recombinant ParasiteSurface Antigen 2 from Leishmania majorinduces a Th1type of immune response but does not protect againstinfection. Vaccine 1998; 16:2077-84; PMID:9796067;DOI:10.1016/S0264-410X(98)00075-9.

    32. Sjlander A, Baldwin TM, Curtis JM, Handman E.

    Induction of a Th1 immune response and simultaneouslack of activation of a Th2 response are required forgeneration of immunity to leishmaniasis. J Immunol1998; 160:3949-57; PMID:9558102.

    33. Handman E, Noormohammadi AH, Curtis JM,Baldwin T, Sjolander A. Therapy of murine cutane-ous leishmaniasis by DNA vaccination. Vaccine 2000;18:3011-7; PMID:10825604; DOI:10.1016/S0264-410X(00)00109-2.

    34. Mougneau E, Altare F, Wakil AE, Zheng S, CoppolaT, Wang ZE, et al. Expression cloning of a protec-tive Leishmania antigen. Science 1995; 268:563-6;PMID:7725103; DOI:10.1126/science.7725103.

    35. Launois P, Maillard I, Pingel S, Swihart KG, XenariosI, Acha-Orbea H, et al. IL-4 rapidly produced by Vbeta 4 V alpha 8 CD4+T cells instructs Th2 develop-ment and susceptibility to Leishmania majorin BALB/cmice. Immunity 1997; 6:541-9; PMID:9175832;DOI:10.1016/S1074-7613(00)80342-8.

    36. Julia V, Rassoulzadegan M, Glaichenhaus N. Resistanceto Leishmania major induced by tolerance to a singleantigen. Science 1996; 274:421-3; PMID:8832890;DOI:10.1126/science.274.5286.421.

    37. Kronenberg K, Brosch S, Butsch F, Tada Y, ShibagakiN, Udey MC, et al. Vaccination with TAT-antigenfusion protein induces protective, CD8(+) T cell-mediated immunity against Leishmania major. J InvestDermatol 2010; 130:2602-10; PMID:20574442;DOI:10.1038/jid.2010.171.

    38. Melby PC, Yang J, Zhao W, Perez LE, Cheng J.Leishmania donovani p36(LACK) DNA vaccineis highly immunogenic but not protective againstexperimental visceral leishmaniasis. Infect Immun2001; 69:4719-25; PMID:11447143; DOI:10.1128/IAI.69.8.4719-25.2001.

    39. Rafati S, Nakhaee A, Taheri T, Taslimi Y, Darabi H,Eravani D, et al. Protective vaccination against experi-mental canine visceral leishmaniasis using a combina-tion of DNA and protein immunization with cysteineproteinases type I and II of L. infantum.Vaccine 2005;23:3716-25; PMID:15882533; DOI:10.1016/j.vac-cine.2005.02.009.

    40. Soong L, Duboise SM, Kima P, McMahon-Pratt D.Leishmania pifanoi amastigote antigens protect miceagainst cutaneous leishmaniasis. Infect Immun 1995;63:3559-66; PMID:7642292.

  • 8/12/2019 KedzierskiHV7-11

    9/11

    1212 Human Vaccines Volume 7 Issue 11

    67. Collin N, Gomes R, Teixeira C, Cheng L,Laughinghouse A, Ward JM, et al. Sand fly sali-vary proteins induce strong cellular immunity in anatural reservoir of visceral leishmaniasis with adverseconsequences for Leishmania. PLoS Pathog 2009;5:1000441; PMID:19461875; DOI:10.1371/journal.ppat.1000441.

    68. Peters NC, Kimblin N, Secundino N, Kamhawi S,Lawyer P, Sacks DL. Vector transmission of leishma-nia abrogates vaccine-induced protective immunity.

    PLoS Pathog 2009; 5:1000484; PMID:19543375;DOI:10.1371/journal.ppat.1000484.

    69. Gomes RB, Brodskyn C, de Oliveira CI, Costa J,Miranda JC, Caldas A, et al. Seroconversion againstLutzomyia longipalpis saliva concurrent with thedevelopment of anti-Leishmania chagasi delayed-type hypersensitivity. J Infect Dis 2002; 186:1530-4;PMID:12404176; DOI:10.1086/344733.

    70. Rohouov I, Hostomska J, Vlkova M, Kobets T,Lipoldova M, Volf P. The protective effect againstLeishmania infection conferred by sand fly bites islimited to short-term exposure. Int J Parasitol 2011;41:481-5; PMID:21310158; DOI:10.1016/j.ijpa-ra.2011.01.003.

    71. Ahmed SB, Kaabi B, Chelbi I, Derbali M, Cherni S,Laouini D, et al. Lack of protection of pre-immuni-zation with saliva of long-term colonized Phlebotomus

    papatasiagainst experimental challenge with Leishmania

    major and saliva of wild-caught P. papatasi. Am JTrop Med Hyg 2010; 83:512-4; PMID:20810812;DOI:10.4269/ajtmh.2010; 09-0687.

    72. Titus RG, Gueiros-Filho FJ, de Freitas LA, BeverleySM. Development of a safe live Leishmania vaccineline by gene replacement. Proc Natl Acad Sci USA1995; 92:10267-71; PMID:7479765; DOI:10.1073/pnas.92.22.10267.

    73. Amaral VF, Teva A, Oliveira-Neto MP, Silva AJ, PereiraMS, Cupolillo E, et al. Study of the safety, immunoge-nicity and efficacy of attenuated and killed Leishmania(Leishmania) majorvaccines in a rhesus monkey (Macacamulatta) model of the human disease. Mem InstOswaldo Cruz 2002; 97:1041-8; PMID:12471434;DOI:10.1590/S0074-02762002000700019.

    74. Alexander J, Coombs GH, Mottram JC. Leishmaniamexicana cysteine proteinase-deficient mutantshave attenuated virulence for mice and potentiate

    a Th1 response. J Immunol 1998; 161:6794-801;PMID:9862710.

    75. Saravia NG, Escorcia B, Osorio Y, Valderrama L,Brooks D, Arteaga L, et al. Pathogenicity and protec-tive immunogenicity of cysteine proteinase-deficientmutants of Leishmania mexicana in non-murine mod-els. Vaccine 2006; 24:4247-59; PMID:16216395;DOI:10.1016/j.vaccine.2005.05.045.

    76. Uzonna JE, Spath GF, Beverley SM, Scott P.Vaccination with phosphoglycan-deficient Leishmaniamajor protects highly susceptible mice from virulentchallenge without inducing a strong Th1 response. JImmunol 2004; 172:3793-7; PMID:15004184.

    77. Kbaer C, Uzonna J, Beverley SM, Scott P.Immunization with persistent attenuated (delta)lpg2Leishmania majorparasites requires adjuvant to provideprotective immunity in C57BL/6 mice. Infect Immun

    2006; 74:777-80; PMID:16369039; DOI:10.1128/IAI.74.1.777-80.2006.

    78. Spth GF, Lye LF, Segawa H, Turco SJ, Beverley SM.Identification of a compensatory mutant (lpg2-REV)of Leishmania major able to survive as amastigotes

    within macrophages without LPG2-dependent glyco-conjugates and its significance to virulence and immu-nization strategies. Infect Immun 2004; 72:3622-7;PMID:15155672; DOI:10.1128/IAI.72.6.3622-7.2004.

    79. Zaph C, Uzonna J, Beverley SM, Scott P. Central mem-ory T cells mediate long-term immunity to Leishmaniamajor in the absence of persistent parasites. Nat Med2004; 10:1104-10; PMID:15448686; DOI:10.1038/nm1108.

    54. Coler RN, Goto Y, Bogatzki L, Raman V, Reed SG.Leish-111f, a recombinant polyprotein vaccine thatprotects against visceral Leishmaniasis by elicitationof CD4+ T cells. Infect Immun 2007; 75:4648-54;PMID:17606603; DOI:10.1128/IAI.00394-07.

    55. Gradoni L, Foglia Manzillo V, Pagano A, Piantedosi D,De Luna R, Gramiccia M, et al. Failure of a multi-sub-unit recombinant leishmanial vaccine (MML) to pro-tect dogs from Leishmania infantum infection and toprevent disease progression in infected animals. Vaccine

    2005; 23:5245-51; PMID:16054272; DOI:10.1016/j.vaccine.2005.07.001.

    56. Trigo J, Abbehusen M, Netto EM, Nakatani M, Pedral-Sampaio G, de Jesus RS, et al. Treatment of caninevisceral leishmaniasis by the vaccine Leish-111f + MPL-SE. Vaccine 2010; 28:3333-40; PMID:20206667;DOI:10.1016/j.vaccine.2010.02.089.

    57. Bertholet S, Goto Y, Carter L, Bhatia A, HowardRF, Carter D, et al. Optimized subunit vaccine pro-tects against experimental leishmaniasis. Vaccine 2009;27:7036-45; PMID:19786136; DOI:10.1016/j.vac-cine.2009.09.066.

    58. Miret J, Nascimento E, Sampaio W, Franca JC,Fujiwara RT, Vale A, et al. Evaluation of an immu-nochemotherapeutic protocol constituted ofN-methyl meglumine antimoniate (Glucantime)and the recombinant Leish-110f + MPL-SE vaccineto treat canine visceral leishmaniasis. Vaccine 2008;

    26:1585-94; PMID:18328956; DOI:10.1016/j.vac-cine.2008.01.026.

    59. Chakravarty J, Kumar S, Trivedi S, Rai VK, Singh A,Ashman JA, et al. A clinical trial to evaluate the safetyand immunogenicity of the LEISH-F1 + MPL-SEvaccine for use in the prevention of visceral leish-maniasis. Vaccine 2011; 29:3531-7; PMID:21414377;DOI:10.1016/j.vaccine.2011.02.096.

    60. Llanos-Cuentas A, Calderon W, Cruz M, Ashman JA,Alves FP, Coler RN, et al. A clinical trial to evaluate thesafety and immunogenicity of the LEISH-F1 + MPL-SE vaccine when used in combination with sodiumstibogluconate for the treatment of mucosal leishmani-asis. Vaccine 2010; 28:7427-35; PMID:20851080;DOI:10.1016/j.vaccine.2010.08.092.

    61. Nascimento E, Fernandes DF, Vieira EP, Campos-Neto A, Ashman JA, Alves FP, et al. A clinicaltrial to evaluate the safety and immunogenicity of

    the LEISH-F1 + MPL-SE vaccine when used incombination with meglumine antimoniate for thetreatment of cutaneous leishmaniasis. Vaccine 2010;28:6581-7; PMID:20688040; DOI:10.1016/j.vac-cine.2010.07.063.

    62. Andrade BB, de Oliveira CI, Brodskyn CI, BarralA, Barral-Netto M. Role of sand fly saliva in humanand experimental leishmaniasis: current insights.Scand J Immunol 2007; 66:122-7; PMID:17635789;DOI:10.1111/j.1365-3083.2007.01964.x.

    63. Kamhawi S, Belkaid Y, Modi G, Rowton E, Sacks D.Protection against cutaneous leishmaniasis resultingfrom bites of uninfected sand flies. Science 2000;290:1351-4; PMID:11082061; DOI:10.1126/sci-ence.290.5495.1351.

    64. Morris RV, Shoemaker CB, David JR, Lanzaro GC,Titus RG. Sandfly maxadilan exacerbates infection with

    Leishmania major and vaccinating against it protectsagainst L. majorinfection. J Immunol 2001; 167:5226-30; PMID:11673536.

    65. Valenzuela JG, Belkaid Y, Garfield MK, Mendez S,Kamhawi S, Rowton ED, et al. Toward a defined anti-Leishmania vaccine targeting vector antigens: charac-terization of a protective salivary protein. J Exp Med2001; 194:331-42; PMID:11489952; DOI:10.1084/

    jem.194.3.331.

    66. Gomes R, Teixeira C, Teixeira MJ, Oliveira F, MenezesMJ, Silva C, et al. Immunity to a salivary protein of asand fly vector protects against the fatal outcome ofvisceral leishmaniasis in a hamster model. Proc Natl

    Acad Sci USA 2008; 105:7845-50; PMID:18509051;DOI:10.1073/pnas.0712153105.

    41. Basu R, Bhaumik S, Basu JM, Naskar K, De T, Roy S.Kinetoplastid membrane protein-11 DNA vaccinationinduces complete protection against both pentavalentantimonial-sensitive and -resistant strains of Leishmaniadonovanithat correlates with inducible nitric oxide syn-thase activity and IL-4 generation: evidence for mixedTh1- and Th2-like responses in visceral leishmaniasis. JImmunol 2005; 174:7160-71; PMID:15905560.

    42. Streit JA, Recker TJ, Donelson JE, Wilson ME. BCGexpressing LCR1 of Leishmania chagasi induces pro-tective immunity in susceptible mice. Exp Parasitol2000; 94:33-41; PMID:10631078; DOI:10.1006/expr.1999.4459.

    43. Stger S, Smith DF, Kaye PM. Immunization witha recombinant stage-regulated surface protein fromLeishmania donovani induces protection against vis-ceral leishmaniasis. J Immunol 2000; 165:7064-71;PMID:11120835.

    44. Tewary P, Jain M, Sahani MH, Saxena S, MadhubalaR. A heterologous prime-boost vaccination regimenusing ORFF DNA and recombinant ORFF proteinconfers protective immunity against experimental vis-ceral leishmaniasis. J Infect Dis 2005; 191:2130-7;PMID:15898000; DOI:10.1086/430348.

    45. Iborra S, Soto M, Carrion J, Nieto A, FernandezE, Alonso C, et al. The Leishmania infantum acidicribosomal protein P0 administered as a DNA vaccineconfers protective immunity to Leishmania majorinfec-

    tion in BALB/c mice. Infect Immun 2003; 71:6562-72; PMID:14573678; DOI:10.1128/IAI.71.11.6562-72.2003.

    46. Saravia NG, Hazbon MH, Osorio Y, Valderrama L,Walker J, Santrich C, et al. Protective immunogenic-ity of the paraflagellar rod protein 2 of Leishmaniamexicana.Vaccine 2005; 23:984-95; PMID:15620471;DOI:10.1016/j.vaccine.2004.07.044.

    47. Aguilar-Be I, da Silva Zardo R, Paraguai de Souza E,Borja-Cabrera GP, Rosado-Vallado M, Mut-MartinM, et al. Cross-protective efficacy of a prophylacticLeishmania donovani DNA vaccine against visceraland cutaneous murine leishmaniasis. Infect Immun2005; 73:812-9; PMID:15664920; DOI:10.1128/IAI.73.2.812-9.2005.

    48. Samant M, Gupta R, Kumari S, Misra P, KhareP, Kushawaha PK, et al. Immunization with theDNA-encoding N-terminal domain of proteophos-

    phoglycan of Leishmania donovani generates Th1-type immunoprotective response against experimentalvisceral leishmaniasis. J Immunol 2009; 183:470-9;PMID:19542458; DOI:10.4049/jimmunol.0900265.

    49. Bhowmick S, Ali N. Identification of novel Leishmaniadonovaniantigens that help define correlates of vaccine-mediated protection in visceral leishmaniasis. PLoSONE 2009; 4:5820; PMID:19503834; DOI:10.1371/

    journal.pone.0005820.

    50. Henriquez FL, Campbell SA, Roberts CW, MullenAB, Burchmore R, Carter KC. Vaccination withrecombinant Leishmania donovani gamma-glutamyl-cysteine synthetase fusion protein protects againstL. donovani infection. J Parasitol 2010; 96:929-36;PMID:20950100; DOI:10.1645/GE-2360.1.

    51. Masih S, Arora SK, Vasishta RK. Efficacy of Leishmaniadonovani ribosomal P1 gene as DNA vaccine in

    experimental visceral leishmaniasis. Exp Parasitol 2011;129:55-64; PMID:21640106; DOI:10.1016/j.exp-para.2011.05.014.

    52. Coler RN, Reed SG. Second-generation vaccinesagainst leishmaniasis. Trends Parasitol 2005; 21:244-9;PMID:15837614; DOI:10.1016/j.pt.2005.03.006.

    53. Skeiky YA, Coler RN, Brannon M, Stromberg E,Greeson K, Crane RT, et al. Protective efficacy of atandemly linked, multi-subunit recombinant leish-manial vaccine (Leish-111f) formulated in MPL adju-vant. Vaccine 2002; 20:3292-303; PMID:12213399;DOI:10.1016/S0264-410X(02)00302-X.

  • 8/12/2019 KedzierskiHV7-11

    10/11

    www.landesbioscience.com Human Vaccines 1213

    107. Resende DM, Caetano BC, Dutra MS, Penido ML,Abrantes CF, Verly RM, et al. Epitope mapping andprotective immunity elicited by adenovirus expressingthe Leishmania amastigotespecific A2 antigen: correla-tion with IFNgamma and cytolytic activity by CD8 +T cells. Vaccine 2008; 26:4585-93; PMID:18588933;DOI:10.1016/j.vaccine.2008.05.091.

    108. Okwor I, Uzonna J. Persistent parasites and immu-nologic memory in cutaneous leishmaniasis: implica-tions for vaccine designs and vaccination strategies.

    Immunol Res 2008; 41:123-36; PMID:18389179;DOI:10.1007/s12026-008-8016-2.

    109. Uzonna JE, Wei G, Yurkowski D, Bretscher P. Immuneelimination of Leishmania major in mice: implica-tions for immune memory, vaccination and reac-tivation disease. J Immunol 2001; 167:6967-74;PMID:11739516.

    110. Birnbaum R, Craft N. Innate immunity and Leishmaniavaccination strategies. Dermatol Clin 2011; 29:89-102;PMID:21095533; DOI:10.1016/j.det.2010.08.014.

    111. Forgber M, Basu R, Roychoudhury K, Theinert S,Roy S, Sundar S, et al. Mapping the antigenicity of theparasites in Leishmania donovaniinfection by proteomeserology. PLoS ONE 2006; 1:40; PMID:17183669;DOI:10.1371/journal.pone.0000040.

    112. Sette A, Rappuoli R. Reverse vaccinology: develop-ing vaccines in the era of genomics. Immunity 2010;33:530-41; PMID:21029963; DOI:10.1016/j.immu-

    ni.2010.09.017.113. Schroeder J, Aebischer T. Vaccines for Leishmaniasis:

    From proteome to vaccine candidates. Hum Vaccin2011; 7:10-5; PMID:21245661; DOI:10.4161/hv.7.0.14556.

    114. Lin HH, Zhang GL, Tongchusak S, Reinherz EL,Brusic V. Evaluation of MHC-II peptide bindingprediction servers: applications for vaccine research.BMC Bioinformatics 2008; 9:22; PMID:19091022;DOI:10.1186/1471-2105-9-S12-S22.

    115. Lin HH, Ray S, Tongchusak S, Reinherz EL, BrusicV. Evaluation of MHC class I peptide binding pre-diction servers: applications for vaccine research.BMC Immunol 2008; 9:8; PMID:18366636;DOI:10.1186/1471-2172-9-8.

    116. Draper SJ, Heeney JL. Viruses as vaccine vectors forinfectious diseases and cancer. Nat Rev Microbiol2010; 8:62-73; PMID:19966816; DOI:10.1038/nrmi-cro2240.

    117. de Goede AL, Boers PH, Dekker LJ, Osterhaus AD,Gruters RA, Rimmelzwaan GF. Characterization ofrecombinant influenza A virus as a vector for HIV-1p17Gag. Vaccine 2009; 27:5735-9; PMID:19647812;DOI:10.1016/j.vaccine.2009.07.032.

    118. Stukova MA, Sereinig S, Zabolotnyh NV, Ferko B,Kittel C, Romanova J, et al. Vaccine potential ofinfluenza vectors expressing Mycobacterium tubercu-losis ESAT-6 protein. Tuberculosis 2006; 86:236-46;PMID:16677861; DOI:10.1016/j.tube.2006.01.010.

    119. Miyahira Y, Garcia-Sastre A, Rodriguez D, RodriguezJR, Murata K, Tsuji M, et al. Recombinant virusesexpressing a human malaria antigen can elicit poten-tially protective immune CD8+responses in mice. ProcNatl Acad Sci USA 1998; 95:3954-9; PMID:9520474;DOI:10.1073/pnas.95.7.3954.

    120. Zheng H, Palese P, Garcia-Sastre A. Antitumor prop-erties of influenza virus vectors. Cancer Res 2000;60:6972-6; PMID:11156398.

    121. Hoffmann E, Krauss S, Perez D, Webby R, WebsterRG. Eight-plasmid system for rapid generation ofinfluenza virus vaccines. Vaccine 2002; 20:3165-70; PMID:12163268; DOI:10.1016/S0264-410X(02)00268-2.

    122. Maassab HF, Bryant ML. The develop-ment of live attenuated cold-adapted influenzavirus vaccine for humans. Rev Med Virol 1999;9:237-44; PMID:10578119; DOI:10.1002/(SICI)1099-1654(199910/12)9:43.0.CO;2-G.

    95. Poot J, Janssen LH, van Kasteren-westerneng TJ, vander Heijden-Liefkens KH, Schijns VE, Heckeroth

    A. Vaccination of dogs with six different candidateleishmaniasis vaccines composed of a chimericalrecombinant protein containing ribosomal and histoneprotein epitopes in combination with different adju-vants. Vaccine 2009; 27:4439-46; PMID:19500553;DOI:10.1016/j.vaccine.2009.05.043.

    96. Bahia D, Gontijo NF, Leon IR, Perales J, Pereira MH,Oliveira G, et al. Antibodies from dogs with canine

    visceral leishmaniasis recognise two proteins from thesaliva of Lutzomyia longipalpis. Parasitol Res 2007;100:449-54; PMID:17058112; DOI:10.1007/s00436-006-0307-8.

    97. Giunchetti RC, Correa-Oliveira R, Martins-FilhoOA, Teixeira-Carvalho A, Roatt BM, de Oliveira

    Aguiar-Soares RD, et al. A killed Leishmania vac-cine with sand fly saliva extract and saponin adju-vant displays immunogenicity in dogs. Vaccine 2008;26:623-38; PMID:18180079; DOI:10.1016/j.vac-cine.2007.11.057.

    98. Dantas-Torres F. Leishmune vaccine: the newest toolfor prevention and control of canine visceral leishmani-osis and its potential as a transmission-blocking vac-cine. Vet Parasitol 2006; 141:1-8; PMID:16750885;DOI:10.1016/j.vetpar.2006.05.001.

    99. Palatnik-de-Sousa CB, Dutra HS, BorojevicR. Leishmania donovani surface glycoconjugate

    GP36 is the major immunogen component of thefucose-mannose ligand (FML). Acta Trop 1993;53:59-72; PMID:8096110; DOI:10.1016/0001-706X(93)90006-W.

    100. da Silva VO, Borja-Cabrera GP, Correia Pontes NN, deSouza EP, Luz KG, Palatnik M, et al. A phase II I trial ofefficacy of the FML-vaccine against canine kala-azar inan endemic area of Brazil (Sao Goncalo do Amaranto,RN). Vaccine 2000; 19:1082-92; PMID:11137242;DOI:10.1016/S0264-410X(00)00339-X.

    101. Borja-Cabrera GP, Correia Pontes NN, da Silva VO,Paraguai de Souza E, Santos WR, Gomes EM, et al.Long lasting protection against canine kala-azar usingthe FML-QuilA saponin vaccine in an endemic area ofBrazil (Sao Goncalo do Amarante, RN). Vaccine 2002;20:3277-84; PMID:12213397; DOI:10.1016/S0264-410X(02)00294-3.

    102. Nogueira FS, Moreira MA, Borja-Cabrera GP, Santos

    FN, Menz I, Parra LE, et al. Leishmune vaccineblocks the transmission of canine visceral leishmani-asis: absence of Leishmania parasites in blood, skinand lymph nodes of vaccinated exposed dogs. Vaccine2005; 23:4805-10; PMID:16011864; DOI:10.1016/j.vaccine.2005.05.011.

    103. Borja-Cabrera GP, Cruz Mendes A, Paraguai de SouzaE, Hashimoto Okada LY, de A Trivellato FA, Kawasaki

    JK, et al. Effective immunotherapy against caninevisceral leishmaniasis with the FML-vaccine. Vaccine2004; 22:2234-43; PMID:15149782; DOI:10.1016/j.vaccine.2003.11.039.

    104. Borja-Cabrera GP, Santos FN, Santos FB, TrivellatoFA, Kawasaki JK, Costa AC, et al. Immunotherapy

    with the saponin enriched-Leishmune vaccine ver-sus immunochemotherapy in dogs with natu-ral canine visceral leishmaniasis. Vaccine 2010;

    28:597-603; PMID:19800443; DOI:10.1016/j.vac-cine.2009.09.071.

    105. Palatnik-de-Sousa CB, Silva-Antunes I, Morgado AdeA, Menz I, Palatnik M, Lavor C. Decrease of the inci-dence of human and canine visceral leishmaniasis afterdog vaccination with Leishmune in Brazilian endemicareas. Vaccine 2009; 27:3505-12; PMID:19464528;DOI:10.1016/j.vaccine.2009.03.045.

    106. Fernandes AP, Costa MM, Coelho EA, Michalick MS,de Freitas E, Melo MN, et al. Protective immunityagainst challenge with Leishmania (Leishmania) cha-

    gasi in beagle dogs vaccinated with recombinant A2protein. Vaccine 2008; 26:5888-95; PMID:18786587;DOI:10.1016/j.vaccine.2008.05.095.

    80. Selvapandiyan A, Debrabant A, Duncan R, Muller J,Salotra P, Sreenivas G, et al. Centrin gene disruptionimpairs stage-specific basal body duplication and cellcycle progression in Leishmania. J Biol Chem 2004;279:25703-10; PMID:15084606; DOI:10.1074/jbc.M402794200.

    81. Selvapandiyan A, Dey R, Nylen S, Duncan R, SacksD, Nakhasi HL. Intracellular replication-deficientLeishmania donovani induces long lasting protectiveimmunity against visceral leishmaniasis. J Immunol2009; 183:1813-20; PMID:19592661; DOI:10.4049/

    jimmunol.0900276.

    82. Kedzierski L, Curtis JM, Doherty PC, Handman E,Kedzierska K. Decreased IL-10 and IL-13 productionand increased CD44 hi T cell recruitment contributeto Leishmania major immunity induced by non-per-sistent parasites. Eur J Immunol 2008; 38:3090-100;PMID:18924210; DOI:10.1002/eji.200838423.

    83. Silvestre R, Cordeiro-Da-Silva A, Santarem N, VergnesB, Sereno D, Ouaissi A. SIR2-deficient Leishmaniainfantum induces a defined IFNgamma/IL-10 pat-tern that correlates with protection. J Immunol 2007;179:3161-70; PMID:17709531.

    84. Breton M, Tremblay MJ, Ouellette M, PapadopoulouB. Live nonpathogenic parasitic vector as a candidatevaccine against visceral leishmaniasis. Infect Immun2005; 73:6372-82; PMID:16177308; DOI:10.1128/IAI.73.10.6372-82.2005.

    85. Lima LV, Carneiro LA, Campos MB, Chagas EJ,Laurenti MD, Corbett CE, et al. Canine visceralleishmaniasis due to Leishmania (L.) infantum cha-gasi in Amazonian Brazil: comparison of the parasitedensity from the skin, lymph node and visceral tissuesbetween symptomatic and asymptomatic, seropositivedogs. Rev Inst Med Trop Sao Paulo 2010; 52:259-66;PMID:21049230.

    86. Postigo JA. Leishmaniasi s in the World HealthOrganization eastern Mediterranean Region. Int J

    Antimicrob Agents 2010; 36:62-5; PMID:20728317;DOI:10.1016/j.ijantimicag.2010.06.023.

    87. Ashford RW. The leishmaniases as emerging andreemerging zoonoses. Int J Parasitol 2000; 30:1269-81; PMID:11113254; DOI:10.1016/S0020-7519(00)00136-3.

    88. Biglino A, Bolla C, Concialdi E, Trisciuoglio A,Romano A, Ferroglio E. Asymptomatic Leishmaniainfantum infection in an area of northwestern Italy(Piedmont region) where such infections are tradition-ally nonendemic. J Clin Microbiol 2010; 48:131-6;PMID:19923480; DOI:10.1128/JCM.00416-09.

    89. Schantz PM, Steurer FJ, Duprey ZH, Kurpel KP, BarrSC, Jackson JE, et al. Autochthonous visceral leishman-iasis in dogs in North America. J Am Vet Med Assoc2005; 226:1316-22; PMID:15844420; DOI:10.2460/

    javma.2005.226.1316.

    90. Ready PD. Leishmaniasis emergence in Europe. EuroSurveill 2010; 15:19505; PMID:20403308.

    91. Baneth G, Shaw SE. Chemotherapy of canine leishmani-osis. Vet Parasitol 2002; 106:315-24; PMID:12079737;DOI:10.1016/S0304-4017(02)00115-2.

    92. Palatnik-de-Sousa CB, dos Santos WR, Franca-SilvaJC, da Costa RT, Reis AB, Palatnik M, et al. Impactof canine control on the epidemiology of canine and

    human visceral leishmaniasis in Brazil. Am J Trop MedHyg 2001; 65:510-7; PMID:11716106.

    93. Courtenay O, Quinnell RJ, Garcez LM, Shaw JJ, DyeC. Infectiousness in a cohort of brazilian dogs: whyculling fails to control visceral leishmaniasis in areasof high transmission. J Infect Dis 2002; 186:1314-20;PMID:12402201; DOI:10.1086/344312.

    94. Reithinger R, Coleman PG, Alexander B, Vieira EP,Assis G, Davies CR. Are insecticide-impregnated dogcollars a feasible alternative to dog culling as a strategyfor controlling canine visceral leishmaniasis in Brazil?Int J Parasitol 2004; 34:55-62; PMID:14711590;DOI:10.1016/j.ijpara.2003.09.006.

  • 8/12/2019 KedzierskiHV7-11

    11/11

    1214 Human Vaccines Volume 7 Issue 11

    143. Dondji B, Perez-Jimenez E, Goldsmith-Pestana K,Esteban M, McMahon-Pratt D. Heterologous prime-boost vaccination with the LACK antigen protectsagainst murine visceral leishmaniasis. Infect Immun2005; 73:5286-9; PMID:16041057; DOI:10.1128/IAI.73.8.5286-9.2005.

    144. Ramiro MJ, Zarate JJ, Hanke T, Rodriguez D,Rodriguez JR, Esteban M, et al. Protection in dogsagainst visceral leishmaniasis caused by Leishmaniainfantum is achieved by immunization with a heter-

    ologous prime-boost regime using DNA and vacciniarecombinant vectors expressing LACK. Vaccine 2003;21:2474-84; PMID:12744881; DOI:10.1016/S0264-410X(03)00032-X.

    145. Tapia E, Perez-Jimenez E, Lopez-Fuertes L, Gonzalo R,Gherardi MM, Esteban M. The combination of DNAvectors expressing IL-12 + IL-18 elicits high protectiveimmune response against cutaneous leishmaniasis afterpriming with DNA-p36/LACK and the cytokines,followed by a booster with a vaccinia virus recombi-nant expressing p36/LACK. Microbes Infect 2003;5:73-84; PMID:12650765; DOI:10.1016/S1286-4579(02)00077-1.

    146. Lange UG, Mastroeni P, Blackwell JM, Stober CB.DNA-Salmonella enterica serovar Typhimuriumprimer-booster vaccination biases towards T helper 1responses and enhances protection against Leishmaniamajorinfection in mice. Infect Immun 2004; 72:4924-

    8; PMID:15271962; DOI:10.1128/IAI.72.8.4924-8.2004.

    147. Pinto EF, Pinheiro RO, Rayol A, Larraga V, Rossi-Bergmann B. Intranasal vaccination against cutaneousleishmaniasis with a particulated leishmanial antigen orDNA encoding LACK. Infect Immun 2004; 72:4521-7; PMID:15271911; DOI:10.1128/IAI.72.8.4521-7.2004.

    148. Rafati S, Baba AA, Bakhshayesh M, Vafa M. Vaccinationof BALB/c mice with Leishmania major amastigote-specific cysteine proteinase. Clin Exp Immunol 2000;120:134-8; PMID:10759774; DOI:10.1046/j.1365-2249.2000.01160.x.

    149. Sukumaran B, Tewary P, Saxena S, Madhubala R.Vaccination with DNA encoding ORFF antigen confersprotective immunity in mice infected with Leishmaniadonovani.Vaccine 2003; 21:1292-9; PMID:12559811;DOI:10.1016/S0264-410X(02)00352-3.

    150. Vlez I D, Gilchrist K, Martinez S, Ramirez-Pineda JR, Ashman JA, Alves FP, et al. Safety andimmunogenicity of a defined vaccine for the pre-vention of cutaneous leishmaniasis. Vaccine 2009;28:329-37; PMID:19879995; DOI:10.1016/j.vac-cine.2009.10.045.

    134. Rivier D, Bovay P, Shah R, Didisheim S, MauelJ. Vaccination against Leishmania major in a CBAmouse model of infection: role of adjuvants andmechanism of protection. Parasite Immunol 1999;21:461-73; PMID:10476055; DOI:10.1046/j.1365-3024.1999.00244.x.

    135. Abdelhak S, Louzir H, Timm J, Blel L, Benlasfar Z,Lagranderie M, et al. Recombinant BCG expressingthe leishmania surface antigen Gp63 induces protec-tive immunity against Leishmania major infectionin BALB/c mice. Microbiology 1995; 141:1585-92;PMID:7551026; DOI:10.1099/13500872-141-7-1585.

    136. Gonzlez CR, Noriega FR, Huerta S, Santiago A , VegaM, Paniagua J, et al. Immunogenicity of a SalmonellatyphiCVD 908 candidate vaccine strain expressing themajor surface protein gp63 of Leishmania mexicanamexicana. Vaccine 1998; 16:1043-52; PMID:9682357;DOI:10.1016/S0264-410X(97)00267-3.

    137. Xu D, Liew FY. Protection against leishmaniasis byinjection of DNA encoding a major surface glycopro-tein, gp63, of L. major.Immunology 1995; 84:173-6;PMID:7750991.

    138. Dumonteil E, Maria Jesus RS, Javier EO, Mariadel Rosario GM. DNA vaccines induce partial pro-tection against Leishmania mexicana. Vaccine 2003;21:2161-8; PMID:12706707; DOI:10.1016/S0264-410X(02)00769-7.

    139. Berberich C, Ramirez-Pineda JR, Hambrecht C, AlberG, Skeiky YA, Moll H. Dendritic cell (DC)-based pro-tection against an intracellular pathogen is dependentupon DC-derived IL-12 and can be induced by molec-ularly defined antigens. J Immunol 2003; 170:3171-9;PMID:12626575.

    140. Ahmed SB, Bahloul C, Robbana C, Askri S, Dellagi K. Acomparative evaluation of different DNA vaccine candi-dates against experimental murine leishmaniasis due toL. major.Vaccine 2004; 22:1631-9; PMID:15068845;DOI:10.1016/j.vaccine.2003.10.046.

    141. Gonzalo RM, Rodriguez JR, Rodriguez D, Gonzalez-Aseguinolaza G, Larraga V, Esteban M. Protectiveimmune response against cutaneous leishmaniasis byprime/booster immunization regimens with vacciniavirus recombinants expressing Leishmania infantump36/LACK and IL-12 in combination with purified p36.Microbes Infect 2001; 3:701-11; PMID:11489418;

    DOI:10.1016/S1286-4579(01)01426-5.

    142. Soussi N, Milon G, Colle JH, Mougneau E,Glaichenhaus N, Goossens PL. Listeria monocytogenesas a short-lived delivery system for the inductionof type 1 cell-mediated immunity against the p36/LACK antigen of Leishmania major. Infect Immun2000; 68:1498-506; PMID:10678966; DOI:10.1128/IAI.68.3.1498-506.2000.

    123. Mishin VP, Nedyalkova MS, Hayden FG, GubarevaLV. Protection afforded by intranasal immuniza-tion with the neuraminidase-lacking mutant ofinfluenza A virus in a ferret model. Vaccine 2005;23:2922-7; PMID:15780741; DOI:10.1016/j.vac-cine.2004.11.058.

    124. Torchilin VP. Recent advances wi th liposomes aspharmaceutical carriers. Nat Rev Drug Discov 2005;4:145-60; PMID:15688077; DOI:10.1038/nrd1632.

    125. Taneichi M, Ishida H, Kajino K, Ogasawara K, Tanaka

    Y, Kasai M, et al. Antigen chemically coupled to thesurface of liposomes are cross-presented to CD8+T cellsand induce potent antitumor immunity. J Immunol2006; 177:2324-30; PMID:16887993.

    126. Doroud D, Zahedifard F, Vatanara A, Najafabadi AR,Rafati S. Cysteine proteinase type I, encapsulated insolid lipid nanoparticles induces substantial protectionagainst Leishmania major infection in C57BL/6 mice.Parasite Immunol 2011; 33:335-48; PMID:21410716;DOI:10.1111/j.1365-3024.2011.01289.x.

    127. Danesh-Bahreini MA, Shokri J, Samiei A, Kamali-Sarvestani E, Barzegar-Jalali M, Mohammadi-SamaniS. Nanovaccine for leishmaniasis: preparation of chi-tosan nanoparticles containing Leishmania superoxidedismutase and evaluation of its immunogenicity inBALB/c mice. Int J Nanomedicine 2011; 6:835-42;PMID:21589651.

    128. Colpitts SL, Dalton NM, Scott P. IL-7 receptor

    expression provides the potential for long-term sur-vival of both CD62Lhigh central memory T cells andTh1 effector cells during Leishmania major infection.

    J Immunol 2009; 182:5702-11; PMID:19380817;DOI:10.4049/jimmunol.0803450.

    129. Mller I, Kropf P, Etges RJ, Louis JA. Gamma interfer-on response in secondary Leishmania major infection:role of CD8+T cells. Infect Immun 1993; 61:3730-8;PMID:8359894.

    130. Darrah PA, Patel DT, De Luca PM, Lindsay RW,Davey DF, Flynn BJ, et al. Multifunctional TH1cells define a correlate of vaccine-mediated protectionagainst Leishmania major. Nat Med 2007; 13:843-50;PMID:17558415; DOI:10.1038/nm1592.

    131. Darrah PA, Hegde ST, Patel DT, Lindsay RW, Chen L,Roederer M, et al. IL-10 production differentially influ-ences the magnitude, quality and protective capacity ofTh1 responses depending on the vaccine platform.

    J Exp Med 2010; 207:1421-33; PMID:20530206;DOI:10.1084/jem.20092532.

    132. Handman E, Button LL, McMaster RW. Leishmaniamajor: production of recombinant gp63, its antigenic-ity and immunogenicity in mice. Exp Parasitol 1990;70:427-35; PMID:2182337; DOI:10.1016/0014-4894(90)90127-X.

    133. Olobo JO, Anjili CO, Gicheru MM, Mbati PA,Kariuki TM, Githure JI, et al. Vaccination of vervetmonkeys against cutaneous leishmaniosis using recom-binant Leishmania major surface glycoprotein (gp63).Vet Parasitol 1995; 60:199-212; PMID:8747903;DOI:10.1016/0304-4017(95)00788-6.