WO2017163171A1 - Chimeric protein, vaccine composition against leishmaniasis, and uses thereof - Google Patents

Chimeric protein, vaccine composition against leishmaniasis, and uses thereof Download PDF

Info

Publication number
WO2017163171A1
WO2017163171A1 PCT/IB2017/051610 IB2017051610W WO2017163171A1 WO 2017163171 A1 WO2017163171 A1 WO 2017163171A1 IB 2017051610 W IB2017051610 W IB 2017051610W WO 2017163171 A1 WO2017163171 A1 WO 2017163171A1
Authority
WO
WIPO (PCT)
Prior art keywords
saponin
chimera
leishmaniasis
protein
sla
Prior art date
Application number
PCT/IB2017/051610
Other languages
French (fr)
Portuguese (pt)
Inventor
Eduardo ANTONIO FERRAZ COELHO
Carlos ALBERTO PEREIRA TAVARES
Vivian TAMIETTI MARTINS
Tiago Antônio De Oliveira MENDES
Mariana COSTA DUARTE
Daniel Menezes SOUZA
Bruno MENDES ROATT
Daniela PAGLIARA LAGE
Original Assignee
Universidade Federal De Minas Gerais - Ufmg
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universidade Federal De Minas Gerais - Ufmg filed Critical Universidade Federal De Minas Gerais - Ufmg
Publication of WO2017163171A1 publication Critical patent/WO2017163171A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/002Protozoa antigens
    • A61K39/008Leishmania antigens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/44Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from protozoa
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a vaccine composition based on a recombinant polypeptide chimera composed of human and mouse CD4 + and CD8 + T-lymphocyte specific epitopes derived from four proteins (LiHypl, LiHyp6, LiHyV and HRF) from Leishmania, which was able to induce protection against visceral and cutaneous leishmaniasis and its use.
  • a recombinant polypeptide chimera composed of human and mouse CD4 + and CD8 + T-lymphocyte specific epitopes derived from four proteins (LiHypl, LiHyp6, LiHyV and HRF) from Leishmania, which was able to induce protection against visceral and cutaneous leishmaniasis and its use.
  • Leishmaniasis are endemic infectious diseases in various tropical and subtropical regions on different continents. It is estimated that 380 million people are at risk of contracting the disease and there is an approximate incidence of 0.5 to 1, 0 and 1, 5 to 2.0 million new cases of visceral leishmaniasis per year ( VL) and cutaneous leishmaniasis (LT), respectively, worldwide. Brazil is the leading VL incidence site in the Americas, accounting for 95% of cases (Alvar J, et al. Leishmaniasis worldwide and global estimates of its incidence. PLoS One. 7: e35671, 2012).
  • an effective vaccine should contain immunogenic antigens conserved on different Leishmania species, and have an affordable cost to the population and their governments.
  • immunogenic antigens conserved on different Leishmania species, and have an affordable cost to the population and their governments.
  • most studies performed use unique antigens, which are expressed in only one of the morphological forms of the parasite (Fernandes AP, et al. Making an anti-amastigote vaccine for visceral leishmaniasis: rational, update and perspectives. Curr Opin Microbiol. 15 : 1-10, 2012).
  • LiHypl XP_001468941 .1
  • LiHyp6 XP_001568689.1
  • HRF CAJ05086.1
  • LiHyV protein (XP_001462854.1) offered recombinant and saponin-associated protection against infection with this same parasite species (Martins VT, et al. Leishmania-specific hypothetical protein expressed in both promastigote and amastigote stages of Leishmania infantum employed for the serodiagnosis of, and as a vaccine candidate against, visceral leishmaniasis (Parasit. Vectors, 8; 363, 2015); thus demonstrating the feasibility of using such antigens as candidate vaccines against LV.
  • Patent document BR132013001 271 entitled Chimeric Protein, Vaccine Composition and Visceral Leishmaniasis Immunodiagnostic Test Kit, refers to a chimeric protein for use in canine and human LV immunodiagnostic vaccine and test composition developed through selection, identification , production and testing of new antigens by analysis proteomics, bioinformatics, peptide synthesis, immunoassay, synthetic gene construction and protein production and purification.
  • Such technology differs from the present invention in that it comprises other immunogenic components which are different from those presented herein.
  • the patent document P 10,609,847, entitled ⁇ Composition Comprising the N-terminal region of Leishmania histone H2B, use thereof for inducing an immune response _ is an immunogenic composition comprising an antigenic peptide consisting of a fragment of histone protein H2B Leishmania is an adjuvant that stimulates the immune response.
  • This technology differs from the present invention in that it comprises only one antigenic compound.
  • the antigen in question differs from any of those described in the present invention.
  • Patent document WO20131 10824 entitled Multi-component ⁇ Chimera for use as a vaccine against infection by Leishmania spp.
  • mammals refers to a chimera, HISA70, composed of different proteins (H2A, H2B, H3, H4, A2 and HSP70) from Leishmania infantum in the composition of a vaccine against Leishmania spp.
  • HISA70 composed of different proteins (H2A, H2B, H3, H4, A2 and HSP70) from Leishmania infantum in the composition of a vaccine against Leishmania spp.
  • Such technology differs from the present invention in that it comprises proteins other than those listed in the present invention.
  • the present invention relates to a chimeric polypeptide vaccine composed of human and mouse CD4 + and CD8 + T-lymphocyte specific epitopes derived from the four proteins (LiHypl, LiHyp6, LiHyV and HRF) that have been described as capable. to induce protection, when administered alone, against L. infantum infection, to induce protection against infection by L. infantum and L. amazonensis, species causing visceral and cutaneous leishmaniasis, respectively, worldwide.
  • the polypeptide chimera has shown promising results in inducing protection against challenge infection with both parasite species and, thus, is a new candidate for protection against distinct Leishmania species.
  • the protection afforded by the chimeric vaccine was superior to that induced by proteins administered alone in the experimental animals with respect to the immunogenicity induced before and after experimental infection, as well as reducing the parasitic load in different organs of the evaluated animals.
  • Figure 1 represents the antigenicity of recombinant chimera.
  • the recombinant chimera (10 ⁇ g, indicated by the black arrow in the figure) was electrophoresed on a 12% SDS-PAGE denaturing gel, stained with bright Coomassie blue G-250 and transferred to a nitrocellulose membrane, which was placed. incubation with the different serum pools. For this, a molecular weight standard was used as marker (A).
  • Reactions with the serum pools of animals immunized with recombinant proteins LiHypl (B), LiHyp6 (C), LiHyV (D), HRF (E) or sera from animals immunized with the chimera itself (F) are shown.
  • Serum pools of mice experimentally infected with L. infantum (G) or L. amazonensis (H) were also used, as well as sera pool from uninfected and unimmunized animals (I), used as negative reaction control.
  • the present invention relates to a polypeptide chimera-containing vaccine composition composed of human and mouse CD4 + and CD8 + T lymphocyte specific epitopes from four Leishmania immunogenic proteins and their use for the protection and / or treatment against leishmaniasis cutaneous and visceral.
  • the leishmaniasis vaccine composition comprises the chimeric protein defined by SEQ ID NO: 1, in addition to the association of pharmaceutically acceptable adjuvants.
  • adjuvants may be selected, without limitation, from those cited in the following literature: Remington's Pharmaceutical Sciences, Mack Publishing, European or Brazilian Pharmacopoeia or new excipients to be preferably saponins.
  • adjuvants should be Th1 immune response inducers, such as saponins, or compounds that are capable of stimulating a cellular immune response prime by the production of IFN- ⁇ and IL-12.
  • the present invention further relates to the use of the chimeric protein in tegumentary and visceral leishmaniasis vaccine compositions.
  • the proposed vaccine composition may be administered by the intradermal, intramuscular, oral, nasal, intravenous, subcutaneous and / or as a device that can be implanted and / or injected.
  • the proteins LiHypl, LiHyp6, LiHyV and HRF were subjected to bioinformatics analysis for selection of immunogenic regions rich in murine and human T lymphocyte epitopes.
  • the Net CTL Pan program was used, so that the best epitopes that bound to the human MHC class I alleles A2, A3 and B7, which together represent over 90% of the human population of any ethnicity, were selected.
  • the same program was used so that epitopes binding to the ⁇ -2-Kd, ⁇ -2-Ld and ⁇ -2-Dd alleles of BALB / c mice were also selected.
  • the epitopes specific to the class I major histocompatibility complex (MHC) molecules that were selected for chimera construction are shown in Tables 1 and 2.
  • the Net MHC II 2.2 program was used to assess the binding affinity of 15 amino acid peptides to the 26 human alleles. Only epitopes with the ability to interact with binding affinity of less than 500 nM and in at least 30% of alleles were selected. The program was also used to select epitopes that bound to the 1-Ad and 1-Ed alleles of BALB / c mice. The epitopes specific to MHC class II molecules that have been selected for chimera construction are shown in Tables 3 and 4. Table 1. In silico prediction of specific epitopes to MHC class I T-lymphocyte molecules from humans selected for recombinant chimera construction.
  • the immunogenicity of recombinant chimera was evaluated in immunized BALB / c mice by assaying IFN- ⁇ , IL-12, GM-CSF, IL-4 and IL-10 cytokines produced by animal splenocytes, 30 days after administration of the last dose of the immunogen.
  • IFN- ⁇ , IL-12, GM-CSF, IL-4 and IL-10 cytokines produced by animal splenocytes 30 days after administration of the last dose of the immunogen.
  • SLA serum-associated cytokines
  • the chimera plus saponin immunized group produced higher levels of IFN- ⁇ , IL-12 and GM-CSF than the levels of these cytokines obtained in the groups immunized with rLiHypl, rLiHyp6, rLiHyV or rHRF plus saponin proteins. No increase was observed in IL-4 and IL-10 production in any of the groups evaluated.
  • the chimera plus saponin immunized group produced higher levels of IFN- ⁇ , IL-12 and GM-CSF than the levels of these cytokines obtained in the groups immunized with rLiHypl, rLiHyp6, rLiHyV or rHRF proteins. saponin.
  • animals in the control groups (saline and saponin) produced significantly higher levels of IL-4 and IL-10 than the other groups evaluated (Table 9).
  • Table 8 Levels of production of IFN- ⁇ , IL-12 and GM-CSF cytokines (pg / ml). Cytokines were quantified in animal splenocyte culture supernatants. The mean ⁇ standard deviation of the groups is shown.
  • Table 9- IL-4 and IL-10 cytokine production levels (in pg / ml). Cytokines were quantified in animal splenocyte culture supernatants. The mean ⁇ standard deviation of the groups is shown.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to a vaccine composition based on a recombinant polypeptide chimera composed of specific epitopes of human and mice CD4+ and CD8+ T lymphocytes derived from four Leishmania proteins (LiHypl, LiHyp6, LiHyV and HRF), capable of inducing a protection against visceral and cutaneous leishmaniasis, and to the use thereof.

Description

PROTEÍNA QUIMÉRICA, COMPOSIÇÃO VACINAL CONTRA  CHEMICAL PROTEIN, VACINAL COMPOSITION AGAINST
LEISHMANIOSES E USOS  Leishmaniasis and Uses
[001 ] A presente invenção refere-se a uma composição vacinai baseada em uma quimera polipeptídica recombinante composta por epitopos específicos de linfócitos T CD4+ e CD8+ de humano e de camundongo derivados de quatro proteínas (LiHypl , LiHyp6, LiHyV e HRF) de Leishmania, que foi capaz de induzir proteção contra a leishmaniose visceral e tegumentar e seu uso. The present invention relates to a vaccine composition based on a recombinant polypeptide chimera composed of human and mouse CD4 + and CD8 + T-lymphocyte specific epitopes derived from four proteins (LiHypl, LiHyp6, LiHyV and HRF) from Leishmania, which was able to induce protection against visceral and cutaneous leishmaniasis and its use.
[002] As leishmanioses são doenças infecciosas endémicas em diversas regiões tropicais e subtropicais em diferentes continentes. Estima-se que 380 milhões de pessoas encontram-se expostas ao risco de contrair a doença e que haja uma incidência aproximada de 0,5 a 1 ,0 e 1 ,5 a 2,0 milhões de novos casos por ano de leishmaniose visceral (LV) e leishmaniose tegumentar (LT), respectivamente, em todo mundo. O Brasil é o principal sítio de incidência de LV no continente americano, sendo responsável por 95% dos casos (Alvar J, et al. Leishmaniasis worldwide and global estimates of its incidence. PLoS One. 7: e35671 , 2012). Leishmaniasis are endemic infectious diseases in various tropical and subtropical regions on different continents. It is estimated that 380 million people are at risk of contracting the disease and there is an approximate incidence of 0.5 to 1, 0 and 1, 5 to 2.0 million new cases of visceral leishmaniasis per year ( VL) and cutaneous leishmaniasis (LT), respectively, worldwide. Brazil is the leading VL incidence site in the Americas, accounting for 95% of cases (Alvar J, et al. Leishmaniasis worldwide and global estimates of its incidence. PLoS One. 7: e35671, 2012).
[003] A gravidade da doença no hospedeiro mamífero, no qual se enquadram o homem e o cão, alcança desde uma lesão cutânea única, no local da picada, até a forma visceral da doença, que pode ser fatal quando na forma aguda e não tratada (Grimaldi, G. e Tesh, R.B. Leishmaniasis of the New World: current concepts and implications for future research. Clin. Microbiol. Rev. 6; 230"50, 1993). Devido à ineficácia das medidas de controle utilizadas, da dificuldade para um diagnóstico correto e dos problemas de toxicidade relacionados aos tratamentos convencionais; o desenvolvimento de vacinas que sejam capazes de induzir imunidade protetora contra a doença torna-se desejável (Tesh RB. Control of zoonotic visceral leishmaniasis: is it time to change strategies? Am J Trop Med Hyg. 52;287-92, 1995). The severity of the disease in the mammalian host, in which both man and dog fall, ranges from a single cutaneous lesion at the stinging site to the visceral form of the disease, which can be fatal when acute and not. (Grimaldi, G. and Tesh, RB Leishmaniasis of the New World: current concepts and implications for future research. Clin. Microbiol. Rev. 6; 230 " 50, 1993). Due to the ineffectiveness of the control measures used, the difficulty For a correct diagnosis and toxicity problems related to conventional treatments, the development of vaccines that are capable of inducing protective immunity against the disease becomes desirable (Tesh RB. Control of zoonotic visceral leishmaniasis: J Trop Med Hyg. 52; 287-92, 1995).
[004] Em modelos murinos, após a cura da leishmaniose causada pela espécie Leishmania major, os animais adquirem imunidade duradoura contra a reinfecção (Afonso LC e Scott P. Immune responses associated with susceptibility of C57BL/10 mice to Leishmania amazonensis. Infect. & Immun. 61 ; 2952"59, 1993). Tal fato tem estimulado o desenvolvimento de pesquisas visando à obtenção de antígenos vacinais que possam ser utilizados como uma medida efetiva de controle da doença. Existem antígenos que vêm sendo avaliados e que preenchem, parcialmente, requisitos para serem empregados como candidatos à vacina (Stãger S, et al. Immunization with a recombinant stage-regulated surface protein from Leishmania donovani induces protection against visceral leishmaniasis. J Immunol. 165; 7064"71 , 2000; Dondji B, et al. Heterologous prime " boost vaccination with the LACK antigen protects against murine visceral leishmaniasis, //7/ecf Immun 73;5286 "89, 2005). [004] In murine models, after the cure of leishmaniasis caused by the species Leishmania major, animals acquire lasting immunity against reinfection (Afonso LC and Scott P. Immune responses associated with susceptibility of C57BL / 10 mice to Leishmania amazonensis. Infect. & Immun 61;. 2952 , "59, 1993). This fact has stimulated the development of research aiming at obtaining vaccine antigens that can be used as an effective disease control measure. There are antigens that have been evaluated and partially meet requirements to be used as vaccine candidates (Stager S, et al. Immunization with a recombinant stage-regulated surface protein from Leishmania donovani induces protection against visceral leishmaniasis. J Immunol. 165; 7064 " 71, 2000; Dondji B, et al. Heterologous prime " boost vaccination with the LACK antigen protects against murine visceral leishmaniasis, // 7 / ecf Immun 73; 5286 " 89, 2005).
[005] Preferencialmente, uma vacina efetiva deveria conter antígenos imunogênicos e conservados em diferentes espécies de Leishmania, além de apresentar um custo acessível à população e seus governos. Entretanto, a maioria dos estudos realizados utiliza antígenos únicos, que são expressos em apenas uma das formas morfológicas do parasita (Fernandes AP, et al. Making an anti-amastigote vaccine for visceral leishmaniasis: rational, update and perspectives. Curr Opin Microbiol. 15:1 -10, 2012). Porém, devido às dificuldades relacionadas à obtenção de antígenos que sejam eficazes contra mais de uma espécie dos parasitas, que sejam de fácil e rápida produção e que não estimulem a produção de anticorpos específicos ao parasita nos animais imunizados, como por outros motivos; ainda não há disponível no mercado uma vacina completamente eficaz no combate às diferentes espécies do parasita Leishmania.  Preferably, an effective vaccine should contain immunogenic antigens conserved on different Leishmania species, and have an affordable cost to the population and their governments. However, most studies performed use unique antigens, which are expressed in only one of the morphological forms of the parasite (Fernandes AP, et al. Making an anti-amastigote vaccine for visceral leishmaniasis: rational, update and perspectives. Curr Opin Microbiol. 15 : 1-10, 2012). However, due to the difficulties related to obtaining antigens that are effective against more than one species of parasites, which are easily and rapidly produced and which do not stimulate the production of parasite-specific antibodies in immunized animals, as for other reasons; A completely effective vaccine against the different species of Leishmania parasite is not yet available on the market.
[006] Em relação à resposta imune, a resistência frente a infecções de espécies de Leishmania está relacionada a um perfil de resposta Th1 , primada pela produção de IFN-γ por macrófagos ativados. Por outro lado, o perfil de resposta Th2, com produção de IL-4 e IL-10, é associado à susceptibilidade à doença (Scott P. Development and regulation of cell-mediated immunity in experimental leishmaniasis. Immunol Res. 27; 489-98, 2003). Diante disso, espécies de Leishmania podem evadir do mecanismo imune através da indução de IL-4 e inibição de IFN-γ nas fases iniciais da infecção. Sendo assim, células T CD4+ e CD8+ são importantes não só para combater e eliminar o parasita do organismo hospedeiro, mas também para a geração de uma memória imunológica voltada ao combate de futuras infecções pelo parasita (Souza VL, et al. Immune and inflammatory responses to Leishmania amazonensis isolated from diferent clinicai forms of human Leishmaniasis in CBA mice. Mem Inst Oswaldo Cruz, v106(1 ):23- 31 , 201 1 ). Regarding the immune response, resistance to infections of Leishmania species is related to a Th1 response profile, which is dominated by IFN-γ production by activated macrophages. On the other hand, the Th2 response profile with IL-4 and IL-10 production is associated with susceptibility to disease (Scott P. Immunol Res. 27; 489- 98, 2003). Given this, Leishmania species can evade the immune mechanism by inducing IL-4 and inhibiting IFN-γ in the early stages of infection. Thus, CD4 + and CD8 + T cells are important not only for fighting and eliminating the parasite from the host organism, but also for generating an immune memory focused on combating future parasite infections (Souza VL, et al. Immune and inflammatory responses to Leishmania amazonensis isolated from different clinical forms of human Leishmaniasis in CBA mice. Mem Inst Oswaldo Cruz, v106 (1): 23- 31, 201 1).
[007] Por meio da utilização da imunoproteômica, recentemente, proteínas de Leishmania infantum foram reconhecidas por anticorpos presentes nos soros de cães com LV sintomática e/ou assintomática (Coelho VT, et al. Identification of proteins in promastigote and amastigote-like Leishmania using an immunoproteomic approach. PLoS Negl Trop Dis. 6(1 ):e1430, 2012). Dentre elas, novos alvos diagnósticos, vacinais e/ou terapêuticos foram identificados. Três dessas proteínas, denominadas LiHypl (XP_001468941 .1 ), LiHyp6 (XP_001568689.1 ) e HRF (CAJ05086.1 ), quando utilizadas sob a forma recombinante e associadas à saponina, como adjuvante, foram capazes de induzir proteção em camundongos BALB/c contra a infecção experimental por L. infantum (Martins VT, et al. Antigenicity, Immunogenicity and Protective Efficacy of Three Proteins Expressed in the Promastigote and Amastigote Stages of Leishmania infantum against Visceral Leishmaniasis. PLoS One. 10(9):e0137683, 2015). Também, em outro estudo realizado, a proteína LiHyV (XP_001462854.1 ) ofertou proteção sob a forma recombinante e associada à saponina, contra a infecção com essa mesma espécie do parasita (Martins VT, et al. A Leishmania- specific hypothetical protein expressed in both promastigote and amastigote stages of Leishmania infantum employed for the serodiagnosis of, and as a vaccine candidate against, visceral leishmaniasis. Parasit. Vectors, 8; 363, 2015); demonstrando assim a factibilidade de utilização de tais antígenos como candidatos vacinas contra a LV.  Through the use of immunoproteomics recently, proteins of Leishmania infantum have been recognized by antibodies present in the sera of dogs with symptomatic and / or asymptomatic VL (Coelho VT, et al. Identification of proteins in promastigote and amastigote-like Leishmania using an immunoproteomic approach (PLoS Negl Trop Dis. 6 (1): e1430, 2012). Among them, new diagnostic, vaccine and / or therapeutic targets were identified. Three of these proteins, called LiHypl (XP_001468941 .1), LiHyp6 (XP_001568689.1) and HRF (CAJ05086.1), when used in recombinant form and associated with saponin as an adjuvant, were able to induce protection in BALB / c mice. against experimental L. infantum infection (Martins VT, et al. Antigenicity, Immunogenicity and Protective Efficacy of Three Proteins Expressed in the Promastigote and Amastigote Stages of Leishmania infantum against Visceral Leishmaniasis. PLoS One. 10 (9): e0137683, 2015) . Also, in another study, LiHyV protein (XP_001462854.1) offered recombinant and saponin-associated protection against infection with this same parasite species (Martins VT, et al. Leishmania-specific hypothetical protein expressed in both promastigote and amastigote stages of Leishmania infantum employed for the serodiagnosis of, and as a vaccine candidate against, visceral leishmaniasis (Parasit. Vectors, 8; 363, 2015); thus demonstrating the feasibility of using such antigens as candidate vaccines against LV.
[008] Atualmente, são encontrados no estado da arte alguns documentos descrevendo o desenvolvimento de antígenos para tratamento de leishmanioses. O documento de patente BR132013001 271 , intitulado Proteína quimérica, composição vacinai e kit para teste imunodiagnóstico de leishmaniose visceral , refere-se a uma proteína quimérica para uso em composição para vacina e teste imunodiagnóstico de LV canina e humana, desenvolvida através da seleção, identificação, produção e teste de novos antígenos por meio de análise proteômica, bioinformática, síntese de peptídeos, imunoensaio, construção de gene sintético e produção e purificação de proteína. Essa tecnologia se difere da presente invenção por compreender outros componentes imunogênicos, os quais são diferentes dos aqui apresentados. [008] Currently, some documents describing the development of antigens for treatment of leishmaniasis are found in the state of the art. Patent document BR132013001 271, entitled Chimeric Protein, Vaccine Composition and Visceral Leishmaniasis Immunodiagnostic Test Kit, refers to a chimeric protein for use in canine and human LV immunodiagnostic vaccine and test composition developed through selection, identification , production and testing of new antigens by analysis proteomics, bioinformatics, peptide synthesis, immunoassay, synthetic gene construction and protein production and purification. Such technology differs from the present invention in that it comprises other immunogenic components which are different from those presented herein.
[009] O documento de patente P 10609847, intitulado ^Composition comprising the n-terminal region of Leishmania histone h2b, use thereof for inducing an immune response _ trata de uma composição imunogênica que compreende um peptídeo antigênico que consiste de fragmento da proteína histona H2B de Leishmania e um adjuvante que estimula a resposta imunológica. Essa tecnologia se difere da presente invenção por compreender apenas um composto antigênico. Além do mais, o antígeno em questão se difere de qualquer um dos descritos na presente invenção. [009] The patent document P 10,609,847, entitled ^ Composition Comprising the N-terminal region of Leishmania histone H2B, use thereof for inducing an immune response _ is an immunogenic composition comprising an antigenic peptide consisting of a fragment of histone protein H2B Leishmania is an adjuvant that stimulates the immune response. This technology differs from the present invention in that it comprises only one antigenic compound. Moreover, the antigen in question differs from any of those described in the present invention.
[0010] O documento de patente WO20131 10824, intitulado ^Multi-component Chimera for use as a vaccine against infection by leishmania spp. In mammals_ refere-se a uma quimera, HISA70, composta por diferentes proteínas (H2A, H2B, H3, H4, A2 e HSP70) de Leishmania infantum na composição de uma vacina contra Leishmania spp. Essa tecnologia se difere da presente invenção por compreender proteínas diferentes das relacionadas na presente invenção. [0010] Patent document WO20131 10824, entitled Multi-component ^ Chimera for use as a vaccine against infection by Leishmania spp. In mammals refers to a chimera, HISA70, composed of different proteins (H2A, H2B, H3, H4, A2 and HSP70) from Leishmania infantum in the composition of a vaccine against Leishmania spp. Such technology differs from the present invention in that it comprises proteins other than those listed in the present invention.
[001 1 ] Dessa forma, a presente invenção se refere a uma vacina quimérica polipeptídica composta por epítopos específicos de linfócitos T CD4+ e CD8+ de humano e camundongo, derivados das quatro proteínas (LiHypl , LiHyp6, LiHyV e HRF) que foram descritas como capazes de induzir proteção, quando administradas isoladamente, contra a infecção por L. infantum, para a indução de proteção contra a infecção pelas espécies L. infantum e L. amazonensis, espécies causadoras de leishmaniose visceral e tegumentar, respectivamente, no mundo. A quimera polipeptídica apresentou resultados promissores na indução de proteção contra a infecção desafio com ambas as espécies de parasitas e, dessa forma, coloca-se como um novo candidato para a proteção contra distintas espécies de Leishmania. A proteção conferida pela vacina quimérica foi superior àquela induzida pelas proteínas administradas isoladamente nos animais de experimentação, no que tange à imunogenicidade induzida antes e após a infecção experimental, bem como na redução da carga parasitária em diferentes órgãos dos animais avaliados. Thus, the present invention relates to a chimeric polypeptide vaccine composed of human and mouse CD4 + and CD8 + T-lymphocyte specific epitopes derived from the four proteins (LiHypl, LiHyp6, LiHyV and HRF) that have been described as capable. to induce protection, when administered alone, against L. infantum infection, to induce protection against infection by L. infantum and L. amazonensis, species causing visceral and cutaneous leishmaniasis, respectively, worldwide. The polypeptide chimera has shown promising results in inducing protection against challenge infection with both parasite species and, thus, is a new candidate for protection against distinct Leishmania species. The protection afforded by the chimeric vaccine was superior to that induced by proteins administered alone in the experimental animals with respect to the immunogenicity induced before and after experimental infection, as well as reducing the parasitic load in different organs of the evaluated animals.
[0012] No estado da técnica, não foi encontrada nenhuma tecnologia desenvolvida para composição vacinai que compreenda os epítopos descritos na presente invenção, o que a torna inovadora e com possiblidade para aplicação industrial. BREVE DESCRIÇÃO DAS FIGURAS  In the state of the art, no technology developed for vaccine composition comprising the epitopes described in the present invention has been found, which makes it innovative and possible for industrial application. BRIEF DESCRIPTION OF THE FIGURES
[0013] A Figura 1 representa a antigenicidade da quimera recombinante. A quimera recombinante (10 μg, indicada pela seta preta na figura) foi submetida à eletroforese em um gel desnaturante de SDS-PAGE a 12%, corada com Coomassie brilhante blue G-250 e transferida para uma membrana de nitrocelulose, a qual foi colocada em incubação com os diferentes pools de soros. Para tal, um padrão de peso molecular foi utilizado como marcador (A). As reações com os pools de soros dos animais imunizados com as proteínas recombinantes LiHypl (B), LiHyp6 (C), LiHyV (D), HRF (E) ou com soros de animais imunizados com a própria quimera (F) são mostradas. Também, foram utilizados pools de soros de camundongos experimentalmente infectados com L. infantum (G) ou L. amazonensis (H), bem como pool de soros de animais não infectados e não imunizados (I), usado como controle negativo da reação.  Figure 1 represents the antigenicity of recombinant chimera. The recombinant chimera (10 μg, indicated by the black arrow in the figure) was electrophoresed on a 12% SDS-PAGE denaturing gel, stained with bright Coomassie blue G-250 and transferred to a nitrocellulose membrane, which was placed. incubation with the different serum pools. For this, a molecular weight standard was used as marker (A). Reactions with the serum pools of animals immunized with recombinant proteins LiHypl (B), LiHyp6 (C), LiHyV (D), HRF (E) or sera from animals immunized with the chimera itself (F) are shown. Serum pools of mice experimentally infected with L. infantum (G) or L. amazonensis (H) were also used, as well as sera pool from uninfected and unimmunized animals (I), used as negative reaction control.
DESCRIÇÃO DETALHADA DA TECNOLOGIA DETAILED DESCRIPTION OF TECHNOLOGY
[0014] A presente invenção refere-se a uma composição vacinai contendo quimera polipeptídica composta por epítopos específicos de linfocitos T CD4+ e CD8+ de humano e camundongo provenientes de quatro proteínas imunogênicas de Leishmania e seu uso para a proteção e/ou tratamento contra as leishmanioses tegumentar e visceral.  The present invention relates to a polypeptide chimera-containing vaccine composition composed of human and mouse CD4 + and CD8 + T lymphocyte specific epitopes from four Leishmania immunogenic proteins and their use for the protection and / or treatment against leishmaniasis cutaneous and visceral.
[0015] Mais especificamente, a proteína quimérica proposta na presente invenção está definida pela SEQ ID No 1 . A composição vacinai contra leishmanioses compreende a proteína quimérica definida pela SEQ ID No 1 , além da associação de adjuvantes farmaceuticamente aceitáveis.  More specifically, the chimeric protein proposed in the present invention is defined by SEQ ID NO: 1. The leishmaniasis vaccine composition comprises the chimeric protein defined by SEQ ID NO: 1, in addition to the association of pharmaceutically acceptable adjuvants.
[0016] Os adjuvantes farmaceuticamente aceitáveis, de acordo com a presente invenção, podem ser selecionados, sem qualquer limitação, dentre aqueles citados na seguinte literatura: Remington's Pharmaceutical Sciences, Mack Publishing, Farmacopéia Européia ou Brasileira ou novos excipientes a serem desenvolvidos, sendo preferencialmente as saponinas. Preferencialmente, os adjuvantes devem ser indutores de resposta imune Th1 , como as saponinas, ou compostos que sejam capazes de estimular uma resposta imune celular primada pela produção de IFN-γ e IL-12. Pharmaceutically acceptable adjuvants according to the present invention may be selected, without limitation, from those cited in the following literature: Remington's Pharmaceutical Sciences, Mack Publishing, European or Brazilian Pharmacopoeia or new excipients to be preferably saponins. Preferably, adjuvants should be Th1 immune response inducers, such as saponins, or compounds that are capable of stimulating a cellular immune response prime by the production of IFN-γ and IL-12.
[0017] A presente invenção ainda refere-se ao uso da proteína quimérica em composições vacinais contra a leishmaniose tegumentar e visceral.  The present invention further relates to the use of the chimeric protein in tegumentary and visceral leishmaniasis vaccine compositions.
[0018] A composição vacinai proposta pode ser administrada pelas vias intradérmica, intramuscular, oral, nasal, intravenosa, subcutânea e/ou como dispositivo que possa ser implantado e/ou injetado. The proposed vaccine composition may be administered by the intradermal, intramuscular, oral, nasal, intravenous, subcutaneous and / or as a device that can be implanted and / or injected.
[0019] A presente invenção pode ser mais bem compreendida através dos exemplos que se seguem, não limitantes.  The present invention may be better understood by the following non-limiting examples.
Exemplo 1 - Construção da quimera recombinante.  Example 1 - Construction of recombinant chimera.
[0020] Para a construção da quimera polipeptídica, as proteínas LiHypl , LiHyp6, LiHyV e HRF foram submetidas a análises de bioinformática para seleção de regiões imunogênicas ricas em epitopos de linfócitos T murinos e humanos. O programa Net CTL Pan foi utilizado, de forma que os melhores epitopos que se ligaram aos alelos de MHC de classe I humano A2, A3 e B7, os quais juntos representam mais de 90% da população humana de qualquer etnia, foram selecionados. O mesmo programa foi utilizado para que epitopos que se liguem aos alelos Η-2-Kd, Η-2-Ld e Η-2-Dd de camundongos BALB/c fossem também selecionados. Os epitopos específicos às moléculas do complexo de histocompatibilidade principal (MHC) de classe I que foram selecionados para a construção da quimera são mostrados nas Tabelas 1 e 2.  For construction of the polypeptide chimera, the proteins LiHypl, LiHyp6, LiHyV and HRF were subjected to bioinformatics analysis for selection of immunogenic regions rich in murine and human T lymphocyte epitopes. The Net CTL Pan program was used, so that the best epitopes that bound to the human MHC class I alleles A2, A3 and B7, which together represent over 90% of the human population of any ethnicity, were selected. The same program was used so that epitopes binding to the Η-2-Kd, Η-2-Ld and Η-2-Dd alleles of BALB / c mice were also selected. The epitopes specific to the class I major histocompatibility complex (MHC) molecules that were selected for chimera construction are shown in Tables 1 and 2.
[0021 ] Com a finalidade de se identificar os epitopos que se liguem ao MHC de classe II, o programa Net MHC II 2.2 foi utilizado, de modo a avaliar a afinidade de ligação de peptídeos de 15 aminoácidos aos 26 alelos do homem. Apenas os epitopos com a capacidade de interagir com uma afinidade de ligação menor que 500 nM e em pelo menos 30% dos alelos foram selecionados. O programa foi também utilizado para selecionar epitopos que se ligaram aos alelos l-Ad e l-Ed de camundongos BALB/c. Os epitopos específicos às moléculas de MHC de classe II que foram selecionados para a construção da quimera são mostrados na Tabela 3 e 4. Tabela 1. Predição in silico de epitopos específicos às moléculas de MHC classe I de linfocitos T de humanos selecionados para construção da quimera recombinante. In order to identify MHC class II binding epitopes, the Net MHC II 2.2 program was used to assess the binding affinity of 15 amino acid peptides to the 26 human alleles. Only epitopes with the ability to interact with binding affinity of less than 500 nM and in at least 30% of alleles were selected. The program was also used to select epitopes that bound to the 1-Ad and 1-Ed alleles of BALB / c mice. The epitopes specific to MHC class II molecules that have been selected for chimera construction are shown in Tables 3 and 4. Table 1. In silico prediction of specific epitopes to MHC class I T-lymphocyte molecules from humans selected for recombinant chimera construction.
MHC classe I de humano  MHC class I of human
Proteína Número Sequência Alelo Net CTL Score  Protein Number Sequence Allele Net CTL Score
1 69-ILNDGRFQL-77 A2 1, 125  1 69-ILNDGRFQL-77 A2 1,125
2 156-MVPDRSVYI-164 A2 1,069  2 156-MVPDRSVYI-164 A2 1,069
LiHypl 3 83 - ASFMPLLER-91 A3 1,090  LiHypl 3 83 - ASFMPLLER-91 A3 1,090
4 77-LPPLPPASF-85 B7 1,398  4 77-LPPLPPASF-85 B7 1,398
5 80-LPPASFMPL-88 B7 1,390  5 80-LPPASFMPL-88 B7 1,390
6 37-SLATAFGLV-45 A2 1,0202  6 37-SLATAFGLV-45 A2 1,0202
LiHypó  LiHypó
7 52-LLYRSTFRH-60 A3 1,2444  7 52-LLYRSTFRH-60 A3 1,2444
8 87-YMAHIRSYM-95 A2 1,0595  8 87-YMAHIRSYM-95 A2 1.0595
9 l l l-FQTNAAAFV-119 A2 1,0653  9 l 1 l-FQTNAAAFV-119 A2 1.0653
HRF 10 88-MAHIRSYMK-96 A3 1, 1722  HRF 10 88-MAHIRSYMK-96 A3 1, 1722
11 112-QTNAAAFVK- 120 A3 1, 1752  11 112-QTNAAAFVK-120 A3 1, 1752
12 115 - A A AF VKKVL- 123 B7 1, 1121  12 115 - A A AF VKKVL- 123 B7 1,121
13 90-SMSMArTTV-98 A2 1,322  13 90-SMSMArTTV-98 A2 1,322
14 69-VSGNGLTIK-77 A3 1,0395  14 69-VSGNGLTIK-77 A3 1.0395
15 83-TPSSARLSM-91 B7 1,7256  15 83-TPSSARLSM-91 B7 1,7256
LiHyV 16 97-TVAQSArTL-105 B7 1, 1648  LiHyV 16 97-TVAQSArTL-105 B7 1, 1648
17 109-MPANSDIRI-117 B7 1, 1793  17 109-MPANSDIRI-117 B7 1, 1793
18 116-RIVATTSSL-124 B7 1,3978  18 116-RIVATTSSL-124 B7 1.3978
19 125 - AP AQSLFDF- 133 B7 1,5079  19 125 - AP AQSLFDF- 133 B7 1.5079
Tabela 2. Predição in silico de epitopos específicos às moléculas de MHC classe I de linfocitos T de camundongos selecionados para construção da quimera recombinante. Table 2. In silico prediction of specific epitopes to mouse T lymphocyte MHC class I molecules selected for recombinant chimera construction.
MHC classe I de camundongo Proteína Número Sequência Alelo Net CTL Score MHC Class I Mouse Protein Number Sequence Allele Net CTL Score
1 77-LPPLPPASF-85 Η-2-Ld 0,27186  1 77-LPPLPPASF-85 Η-2-Ld 0.27186
2 79-PLPPASFMP-87 Η-2-Ld 0,08248  2 79-PLPPASFMP-87 Η-2-Ld 0.08248
LiHypl 3 165-MSGPARYV Y- 173 Η-2-Dd 0,23280  LiHypl 3 165-MSGPARYV Y-173 Η-2-Dd 0.23280
4 59-DVYTRASDR-67 Η-2-Kd 0,07271  4 59-DVYTRASDR-67 Η-2-Kd 0.07271
5 83 - ASFMPLLER-91 Η-2-Kd 0,20710  5 83 - ASFMPLLER-91 Η-2-Kd 0.20710
LiHypó 7 29-LTYAETVVS-37 Η-2-Kd - LiHypó 7 29-LTYAETVVS-37 Η-2-Kd -
LiHyV 8 82-STPSSARLS-90 Η-2-Ld 0, 11316 LiHyV 8 82-STPSSARLS-90 Η-2-Ld 0, 11316
Tabela 3. Predição in silico de epitopos específicos às moléculas de MHC classe II de linfocitos T de humanos selecionados para construção da quimera recombinante. Table 3. In silico prediction of epitopes specific to MHC class II T-lymphocyte molecules from humans selected for recombinant chimera construction.
MHC classe II de humano  MHC class II of human
Proteína Número Sequência Net MHCII Score  Protein Number Sequence Net MHCII Score
1 52-LLYRSTFRHAMLLRV-66 38,46  1 52-LLYRSTFRHAMLLRV-66 38.46
2 53-LYRSTFRHAMLLRVQ-67 38,46  2 53-LYRSTFRHAMLLRVQ-67 38.46
LiHypó  LiHypó
3 54-YRSTFRHAMLLRVQR-68 38,46  3 54-YRSTFRHAMLLRVQR-68 38.46
4 55-RSTFRHAMLLRVQRE-69 42,31  4 55-RSTFRHAMLLRVQRE-69 42.31
5 56-STFRHAMLLRVQRET-70 38,46  5 56-STFRHAMLLRVQRET-70 38.46
HRF 6 108 -RKAFQTN A A AF VKKV - 122 30,77  HRF 6 108 -RKAFQTN A A AF VKKV - 122 30.77
Tabela 4. Predição in silico de epitopos específicos às moléculas de MHC classe II de linfocitos T de camundongos selecionados para construção da quimera recombinante. Table 4. In silico prediction of specific epitopes to mouse T lymphocyte MHC class II molecules selected for recombinant chimera construction.
MHC classe II de camundongo  MHC Class II Mouse
Proteína Número Sequência Alelo Net MHCII Score  Protein Number Sequence Allele Net MHCII Score
1 169- ARYV YFHMVLPVEAQ- 183 Η-2-IAd 394,4  1 169- ARYV YFHMVLPVEAQ- 183 Η-2-IAd 394.4
LiHypl  LiHypl
2 170-RYV YFHMVLPVEAQR- 184 Η-2-IAd 446,9 3 171 - YV YFHMVLPVEAQRF- 185 Η-2-IAd 161,2 2 170-RYV YFHMVLPVEAQR-184 Η-2-IAd 446.9 3 171 - YV YFHMVLPVEAQRF- 185 Η-2-IAd 161,2
4 172- V YFHMVLPVEAQRFS - 186 Η-2-IAd 160,5  4 172- V YFHMVLPVEAQRFS - 186 Η-2-IAd 160.5
5 173- YFHMVLPVEAQRFSL- 187 Η-2-IAd 169,2  5 173- YFHMVLPVEAQRFSL-187 Η-2-IAd 169.2
6 174-FHMVLPVEAQRFSLV- 188 Η-2-IAd 255,3  6 174-FHMVLPVEAQRFSLV-188 Η-2-IAd 255.3
7 55-RSTFRHAMLLRVQRE-69 Η-2-IAd 204,8  7 55-RSTFRHAMLLRVQRE-69 Η-2-IAd 204.8
8 56-STFRHAMLLRVQRET-70 Η-2-IAd 182,5  8 56-STFRHAMLLRVQRET-70 Η-2-IAd 182.5
LiHypó 9 57-TFRHAMLLRVQRETR-71 Η-2-IAd 221,1  LiHypó 9 57-TFRHAMLLRVQRETR-71 Η-2-IAd 221.1
10 58 -FRH AMLLR VQRETRF-72 Η-2-IAd 252,0  10 58 -FRH AMLLR VQRETRF-72 Η-2-IAd 252.0
11 59-RHAMLLRVQRETRFD-73 Η-2-IAd 484,1  11 59-RHAMLLRVQRETRFD-73 Η-2-IAd 484.1
12 83-TPSSARLSMSMArfT-97 Η-2-IAd 411,1  12 83-TPSSARLSMSMArfT-97 Η-2-IAd 411.1
13 84-PSSARLSMSMArfTV-98 Η-2-IAd 347,9  13 84-PSSARLSMSMArfTV-98 Η-2-IAd 347.9
14 85-SSARLSMSMArTTVA-99 Η-2-IAd 193,3  14 85-SSARLSMSMArTTVA-99 Η-2-IAd 193.3
15 86-SARLSMSMArrTVAQ- 100 Η-2-IAd 145,8  15 86-SARLSMSMArrTVAQ- 100 Η-2-IAd 145.8
16 87-ARLSMSMArTTV AQS- 101 Η-2-IAd 113,9  16 87-ARLSMSMArTTV AQS-101 Η-2-IAd 113.9
LiHyV 17 88-RLSMSMArTTVAQSA-102 Η-2-IAd 93,5  LiHyV 17 88-RLSMSMArTTVAQSA-102 Η-2-IAd 93.5
18 89-LSMSMAnTVAQSAI-103 Η-2-IAd 165,8  18 89-LSMSMAnTVAQSAI-103 Η-2-IAd 165.8
19 90-SMSMArTTVAQSArr-104 Η-2-IAd 231,5  19 90-SMSMArTTVAQSArr-104 Η-2-IAd 231.5
20 97-TVAQSArTLSGVMPA-l 11 Η-2-IAd 382,5  20 97-TVAQSArTLSGVMPA-l 11 Η-2-IAd 382.5
21 98-VAQSAITLSGVMPAN-l 12 Η-2-IAd 391,0  21 98-VAQSAITLSGVMPAN-l 12 Η-2-IAd 391.0
Exemplo 2 - Validação da antigenicidade da quimera. Example 2 - Validation of chimera antigenicity.
[0022] Com o objetivo de validar a antigenicidade da quimera polipeptídica, experimentos de Western-blot foram realizados utilizando soros de camundongos BALB/c que foram imunizados por via subcutânea com as proteínas rLiHypl , rl_iHyp6, rLiHyV ou rHRF mais saponina, bem como com soros de animais cronicamente infectados com L. infantum ou L. amazonensis. Nos resultados, pode-se observar que todos os pools de soros utilizados (n=8, por grupo), com exceção do controle negativo, que foi composto por soros de animais que não foram imunizados ou infectados; foram capazes de reconhecer a quimera polipeptídica (-30.0 kDa), demonstrando assim a antigenicidade da proteína recombinante nos animais vacinados e/ou infectados (Figura 1 ). Exemplo 3- Avaliação da resposta imune celular induzida pela quimera antes da infecção desafio. In order to validate the antigenicity of the polypeptide chimera, Western blot experiments were performed using BALB / c mouse sera that were subcutaneously immunized with the rLiHypl, rl_iHyp6, rLiHyV or rHRF plus saponin proteins. sera from animals chronically infected with L. infantum or L. amazonensis. In the results, it can be observed that all serum pools used (n = 8, per group), except for the negative control, which was composed of sera from animals that were not immunized or infected; were able to recognize polypeptide chimera (-30.0 kDa), thus demonstrating the antigenicity of the recombinant protein in vaccinated and / or infected animals (Figure 1). Example 3- Assessment of chimera-induced cellular immune response prior to challenge infection.
[0023] A imunogenicidade da quimera recombinante foi avaliada em camundongos BALB/c imunizados, por meio da dosagem das citocinas IFN-γ, IL- 12, GM-CSF, IL-4 e IL-10 produzidas pelos esplenocitos dos animais, 30 dias após a administração da última dose do imunógeno. Após o estímulo in vitro com as proteínas específicas ou com uma mistura de extratos dos dois parasitas (SLA), observou-se a produção de níveis significativamente elevados de IFN-γ, IL- 12 e GM-CSF no grupo dos animais vacinados, em relação aos níveis produzidos pelos esplenocitos dos grupos controle, constituídos por animais que receberam saponina ou salina (Tabela 5). Também, o grupo imunizado com a quimera mais saponina produziu maiores níveis de IFN-γ, IL-12 e GM-CSF em relação aos níveis dessas citocinas obtidos nos grupos imunizados com as proteínas rLiHypl , rLiHyp6, rLiHyV ou rHRF mais saponina. Nenhum aumento foi observado na produção de IL-4 e IL-10 em qualquer dos grupos avaliados.  The immunogenicity of recombinant chimera was evaluated in immunized BALB / c mice by assaying IFN-γ, IL-12, GM-CSF, IL-4 and IL-10 cytokines produced by animal splenocytes, 30 days after administration of the last dose of the immunogen. Following in vitro stimulation with specific proteins or a mixture of extracts of the two parasites (SLA), significantly elevated levels of IFN-γ, IL-12 and GM-CSF were observed in the vaccinated group of animals. regarding the levels produced by the splenocytes of the control groups, consisting of animals that received saponin or saline (Table 5). Also, the chimera plus saponin immunized group produced higher levels of IFN-γ, IL-12 and GM-CSF than the levels of these cytokines obtained in the groups immunized with rLiHypl, rLiHyp6, rLiHyV or rHRF plus saponin proteins. No increase was observed in IL-4 and IL-10 production in any of the groups evaluated.
Tabela 5- Níveis de produção das citocinas IFN-γ, IL-12 e GM-CSF (em pg/ml). As citocinas foram quantificadas em sobrenadantes de cultura de esplenocitos dos animais. A média ± desvio padrão dos grupos é mostrada. Table 5- Production levels of IFN-γ, IL-12 and GM-CSF cytokines (in pg / ml). Cytokines were quantified in animal splenocyte culture supernatants. The mean ± standard deviation of the groups is shown.
Citocinas (em pg/ml)  Cytokines (in pg / ml)
Estímulo IFN-gama IL-12 GM-CSF  IFN-gamma stimulation IL-12 GM-CSF
Meio 66+8 55+5 60+5 Medium 66 + 8 55 + 5 60 + 5
Salina Mix proteínas 70+10 67+8 57+7 Saline Protein Mix 70 + 10 67 + 8 57 + 7
SLA 58+6 69+6 59+8  SLA 58 + 6 69 + 6 59 + 8
Meio 64+5 55+4 70+8Medium 64 + 5 55 + 4 70 + 8
Saponina Mix proteínas 77+10 65+8 77+6 Saponin Protein Mix 77 + 10 65 + 8 77 + 6
SLA 59+10 59+7 59+5  SLA 59 + 10 59 + 7 59 + 5
Meio 56+5 58+9 64+4 LiHyp 1/saponina LiHypl 998+98 233+55 199+33 Middle 56 + 5 58 + 9 64 + 4 LiHyp 1 / LiHypl saponin 998 + 98 233 + 55 199 + 33
SLA 433+35 155+22 138+16  SLA 433 + 35 155 + 22 138 + 16
Meio 55+5 57+7 63+6Medium 55 + 5 57 + 7 63 + 6
LiHypó/saponina LiHYpó 677+57 201+21 177+26 LiHypó / saponin LiHYpo 677 + 57 201 + 21 177 + 26
SLA 322+20 134+15 107+10  SLA 322 + 20 134 + 15 107 + 10
Meio 60+10 64+4 58+5Medium 60 + 10 64 + 4 58 + 5
LiHyV/saponina LiHyV 905+45 195+30 180+30 LiHyV / Saponin LiHyV 905 + 45 195 + 30 180 + 30
SLA 405+45 133+17 121+19  SLA 405 + 45 133 + 17 121 + 19
Meio 49+5 54+5 62+7Medium 49 + 5 54 + 5 62 + 7
HRF/saponina HRF 877+55 178+20 160+22 HRF / saponin HRF 877 + 55 178 + 20 160 + 22
SLA 377+32 125+20 121+17  SLA 377 + 32 125 + 20 121 + 17
Meio 59+9 63+5 58+6Medium 59 + 9 63 + 5 58 + 6
Quimera/saponina Quimera 1.644+109 425+40 307+32 Chimera / Saponin Chimera 1,644 + 109 425 + 40 307 + 32
SLA 766+30 255+20 170+15  SLA 766 + 30 255 + 20 170 + 15
Exemplo 4- Avaliação da eficácia de proteção contra a infecção desafio por L. infantum ou L. amazonensis. Example 4- Evaluation of protective efficacy against challenge infection by L. infantum or L. amazonensis.
[0024] Dez semanas após a infecção desafio, a carga parasitária dos diversos grupos foi avaliada na pata infectada (L amazonensis), no fígado, baço, linfonodo drenante e medula óssea dos animais infectados com L. infantum (Tabela 6) ou L. amazonensis (Tabela 7). Pode-se observar que, em ambos os casos, ainda que os animais imunizados com as proteínas recombinantes individuais mais saponina tenham apresentado menor carga parasitária em relação aos grupos controles (salina e saponina), os animais imunizados com a quimera recombinante mais saponina apresentaram reduções mais significativas no parasitismo quando comparado a todos os demais grupos experimentais. Ten weeks after challenge infection, the parasitic burden of the various groups was evaluated on the infected paw (L amazonensis), liver, spleen, draining lymph node and bone marrow of animals infected with L. infantum (Table 6) or L. amazonensis (Table 7). It can be observed that in both cases, although animals immunized with individual recombinant proteins plus saponin had lower parasitic load than control groups (saline and saponin), animals immunized with the most recombinant chimera saponin showed more significant reductions in parasitism when compared to all other experimental groups.
Tabela 6. Resultados da carga parasitária dos animais imunizados e desafiados com L. infantum. A carga parasitária foi determinada por uma técnica de diluição limitante, 10 semanas após a infecção desafio. A média ± desvio padrão dos grupos é mostrada.  Table 6. Results of parasitic burden of animals immunized and challenged with L. infantum. Parasitic load was determined by a limiting dilution technique 10 weeks after challenge infection. The mean ± standard deviation of the groups is shown.
Figure imgf000014_0001
Figure imgf000014_0001
Tabela 7. Resultados da carga parasitária dos animais imunizados e desafiados com L. amazonensis. A carga parasitária foi determinada por uma técnica de diluição limitante, 10 semanas após a infecção desafio. A média ± desvio padrão dos grupos é mostrada. Table 7. Results of parasitic load of animals immunized and challenged with L. amazonensis. Parasitic load was determined by a limiting dilution technique 10 weeks after challenge infection. The mean ± standard deviation of the groups is shown.
Pata infectada Baço Fígado Linfonodos Medula óssea Infected Paw Spleen Liver Lymph Nodes Bone Marrow
Salina 8,5+0,5 6,5+0,4 4,4+0,2 7,3+0,4 3,3+0,2Saline 8.5 + 0.5 6.5 + 0.4 4.4 + 0.2 7.3 + 0.4 3.3 + 0.2
Saponina 8,0+0,3 6,1+0,2 4,1+0,4 6,8+0,5 3,1+0,3Saponin 8.0 + 0.3 6.1 + 0.2 4.1 + 0.4 6.8 + 0.5 3.1 + 0.3
LiHyp 1/saponina 3,2+0,3 2,7+0,4 2,5+0,3 3,1+0,3 1,9+0,3LiHyp 1 / saponin 3.2 + 0.3 2.7 + 0.4 2.5 + 0.3 3.1 + 0.3 1.9 + 0.3
LiHypó/saponina 3,7+0,4 3,1+0,3 2,9+0,4 3,7+0,3 2,3+0,3LiHypó / saponin 3.7 + 0.4 3.1 + 0.3 2.9 + 0.4 3.7 + 0.3 2.3 + 0.3
LiHyV/saponina 3,0+0,3 2,6+0,2 2,3+0,4 3,0+0,2 1,9+0,5LiHyV / saponin 3.0 + 0.3 2.6 + 0.2 2.3 + 0.4 3.0 + 0.2 1.9 + 0.5
HRF/saponina 4,1+0,4 3,7+0,4 3,3+0,4 3,9+0,3 2,5+0,4HRF / saponin 4.1 + 0.4 3.7 + 0.4 3.3 + 0.4 3.9 + 0.3 2.5 + 0.4
Quimera/saponina 1,8+0,3 1,6+0,2 1,5+0,3 1,6+0,5 0,8+0,3 Exemplo 5- Avaliação da resposta imune celular após a infecção desafio. Chimera / saponin 1.8 + 0.3 1.6 + 0.2 1.5 + 0.3 1.6 + 0.5 0.8 + 0.3 Example 5- Evaluation of cellular immune response following challenge infection.
[0025] A avaliação da resposta imune celular após a infecção desafio foi também realizada. As culturas de esplenócitos dos animais imunizados e infectados foram estimuladas com as proteínas específicas usadas nas imunizações ou com uma mistura de extratos dos dois parasitas (SLA) e procederam-se às dosagens das citocinas IFN-γ, IL-12, GM-CSF, IL-4 e IL-10. Nos resultados, observou-se a produção de níveis significativamente elevados de IFN-γ, IL-12 e GM-CSF no grupo dos animais vacinados, em relação aos níveis produzidos dessas citocinas nos esplenócitos dos grupos controle (Tabela 8). Como observado antes da infecção, o grupo imunizado com a quimera mais saponina produziu maiores níveis de IFN-γ, IL-12 e GM-CSF em relação aos níveis dessas citocinas obtidos nos grupos imunizados com as proteínas rLiHypl , rLiHyp6, rLiHyV ou rHRF mais saponina. Entretanto, os animais dos grupos controles (salina e saponina) produziram níveis significativamente maiores de IL-4 e IL-10, em relação aos demais grupos avaliados (Tabela 9).  Evaluation of cellular immune response following challenge infection was also performed. Splenocyte cultures of the immunized and infected animals were stimulated with the specific proteins used in the immunizations or a mixture of extracts from the two parasites (SLA) and the IFN-γ, IL-12, GM-CSF, IL-4 and IL-10. Results showed significantly higher levels of IFN-γ, IL-12 and GM-CSF in the vaccinated group compared to the levels of these cytokines in the splenocytes of the control groups (Table 8). As observed before infection, the chimera plus saponin immunized group produced higher levels of IFN-γ, IL-12 and GM-CSF than the levels of these cytokines obtained in the groups immunized with rLiHypl, rLiHyp6, rLiHyV or rHRF proteins. saponin. However, animals in the control groups (saline and saponin) produced significantly higher levels of IL-4 and IL-10 than the other groups evaluated (Table 9).
Tabela 8- Níveis de produção das citocinas IFN-γ, IL-12 e GM-CSF ( pg/ml). As citocinas foram quantificadas em sobrenadantes de cultura esplenócitos dos animais. A média ± desvio padrão dos grupos é mostrada. Table 8- Levels of production of IFN-γ, IL-12 and GM-CSF cytokines (pg / ml). Cytokines were quantified in animal splenocyte culture supernatants. The mean ± standard deviation of the groups is shown.
Citocinas (em pg/ml)  Cytokines (in pg / ml)
Estímulo IFN-gama IL-12 GM-CSF  IFN-gamma stimulation IL-12 GM-CSF
Meio 77+10 66+5 57+7 Medium 77 + 10 66 + 5 57 + 7
Salina Mix proteínas 88+7 78+7 68+8 Saline Protein Mix 88 + 7 78 + 7 68 + 8
SLA 80+5 88+9 79+9  SLA 80 + 5 88 + 9 79 + 9
Meio 66+4 68+3 58+4Medium 66 + 4 68 + 3 58 + 4
Saponina Mix proteínas 96+9 88+6 77+8 Saponin Mix Proteins 96 + 9 88 + 6 77 + 8
SLA 101+15 99+7 88+4 Meio 66+7 65+4 58+6SLA 101 + 15 99 + 7 88 + 4 Medium 66 + 7 65 + 4 58 + 6
LiHyp 1/saponina LiHyp 1 1.211+105 343+35 266+25 LiHyp 1 / saponin LiHyp 1 1,211 + 105 343 + 35 266 + 25
SLA 776+50 257+28 199+20  SLA 776 + 50 257 + 28 199 + 20
Meio 60+4 55+4 58+4Medium 60 + 4 55 + 4 58 + 4
LiHypó/saponina LiHYpó 977+46 255+33 199+20 LiHypó / saponina LiHYpo 977 + 46 255 + 33 199 + 20
SLA 554+35 199+21 166+22  SLA 554 + 35 199 + 21 166 + 22
Meio 48+7 55+6 63+3Medium 48 + 7 55 + 6 63 + 3
LiHyV/saponina LiHyV 1.101+90 288+23 201+26 LiHyV / LiHyV saponin 1,101 + 90 288 + 23,203 + 26
SLA 667+33 255+20 196+25  SLA 667 + 33 255 + 20 196 + 25
Meio 52+2 59+6 55+4Medium 52 + 2 59 + 6 55 + 4
HRF/saponina HRF 933+25 233+18 177+19 HRF / saponin HRF 933 + 25 233 + 18 177 + 19
SLA 477+23 177+21 155+10  SLA 477 + 23 177 + 21 155 + 10
Meio 63+6 65+7 68+8Medium 63 + 6 65 + 7 68 + 8
Quimera/saponina Quimera 2.322+122 544+30 372+20 Chimera / Saponin Chimera 2,322 + 122,544 + 30,372 + 20
SLA 1.556+99 388+35 295+45  SLA 1.556 + 99 388 + 35 295 + 45
Tabela 9- Níveis de produção das citocinas IL-4 e IL-10 (em pg/ml). As citocinas foram quantificadas em sobrenadantes de cultura de esplenocitos dos animais. A média ± desvio padrão dos grupos é mostrada. Table 9- IL-4 and IL-10 cytokine production levels (in pg / ml). Cytokines were quantified in animal splenocyte culture supernatants. The mean ± standard deviation of the groups is shown.
Citocinas (em pg/ml)  Cytokines (in pg / ml)
Estímulo IL-4 IL-10  IL-4 Stimulus IL-10
Meio 59+7 65+6  Medium 59 + 7 65 + 6
Salina Mix proteínas 98+12 86+5 SLA 488+40 455+25 Saline Protein Mix 98 + 12 86 + 5 SLA 488 + 40 455 + 25
Meio 64+5 55+4Medium 64 + 5 55 + 4
Saponina Mix proteínas 77+10 65+8 Saponin Protein Mix 77 + 10 65 + 8
SLA 356+88 367+48  SLA 356 + 88 367 + 48
Meio 77+8 68+10Medium 77 + 8 68 + 10
LiHyp 1/saponina LiHyp 1 70+8 67+4 LiHyp 1 / saponin LiHyp 1 70 + 8 67 + 4
SLA 79+10 83+4  SLA 79 + 10 83 + 4
Meio 65+6 68+8Medium 65 + 6 68 + 8
LiHypó/saponina LiHYpó 77+5 77+4 LiHypó / Saponina LiHY Powder 77 + 5 77 + 4
SLA 80+10 86+9  SLA 80 + 10 86 + 9
Meio 59+7 66+7Medium 59 + 7 66 + 7
LiHyV/saponina LiHyV 70+5 75+10 LiHyV / LiHyV Saponin 70 + 5 75 + 10
SLA 88+5 87+10  SLA 88 + 5 87 + 10
Meio 70+6 68+8Medium 70 + 6 68 + 8
HRF/saponina HRF 121+11 133+25 HRF / saponin HRF 121 + 11 133 + 25
SLA 101+9 106+12  SLA 101 + 9 106 + 12
Meio 55+6 59+4Medium 55 + 6 59 + 4
Quimera/saponina Quimera 66+5 75+6 Chimera / Saponin Chimera 66 + 5 75 + 6
SLA 73+7 80+6  SLA 73 + 7 80 + 6

Claims

REIVINDICAÇÕES
1 - PROTEÍNA QUIMÉRICA, caracterizada por compreender a sequência polipeptídica definida pela SEQ ID N° 1 . 1 - CHEMICAL PROTEIN, characterized in that it comprises the polypeptide sequence defined by SEQ ID NO: 1.
2- COMPOSIÇÃO VACINAL contra leishmaniose tegumentar e visceral, caracterizada por compreender a proteína quimérica definida na reivindicação 1 e adjuvantes farmaceuticamente aceitáveis.  VACINAL COMPOSITION against cutaneous and visceral leishmaniasis, characterized in that it comprises the chimeric protein defined in claim 1 and pharmaceutically acceptable adjuvants.
3- COMPOSIÇÃO VACINAL contra leishmaniose tegumentar e visceral, de acordo com a reivindicação 2, caracterizada pelos adjuvantes serem, preferencialmente, compostos capazes de induzir ao desenvolvimento de uma resposta imunológica do tipo Th1 , primada pela produção das citocinas IFN-γ e IL- 12, como a saponina.  VACINAL COMPOSITION against tegumentary and visceral leishmaniasis according to claim 2, characterized in that the adjuvants are preferably compounds capable of inducing the development of a Th1-type immune response, primared by the production of IFN-γ and IL-12 cytokines. , like the saponin.
4- COMPOSIÇÃO VACINAL contra leishmaniose tegumentar e visceral, de acordo com a reivindicação 2, caracterizada por poder ser administrada pelas vias intradérmica, intramuscular, oral, nasal, intravenosa, subcutânea e/ou como dispositivo que possa ser implantado e/ou injetado.  VACINAL COMPOSITION against cutaneous and visceral leishmaniasis according to claim 2, characterized in that it can be administered by the intradermal, intramuscular, oral, nasal, intravenous, subcutaneous routes and / or as a device that can be implanted and / or injected.
5- USO DA PROTEÍNA QUIMÉRICA definida na reivindicação 1 , caracterizado por ser para o tratamento e/ou prevenção das leishmanioses tegumentar e visceral em cães e humanos.  Use of the CHEMERIC PROTEIN defined in claim 1, characterized in that it is for the treatment and / or prevention of cutaneous and visceral leishmaniasis in dogs and humans.
6- USO DA COMPOSIÇÃO VACINAL definida nas reivindicações 2 a 4, caracterizado por ser para o tratamento e/ou prevenção das leishmanioses tegumentar e visceral em cães e humanos.  USE OF THE VACINAL COMPOSITION as defined in claims 2 to 4, characterized in that it is for the treatment and / or prevention of cutaneous and visceral leishmaniasis in dogs and humans.
PCT/IB2017/051610 2016-03-21 2017-03-20 Chimeric protein, vaccine composition against leishmaniasis, and uses thereof WO2017163171A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
BR102016006121-0A BR102016006121A2 (en) 2016-03-21 2016-03-21 CHEMICAL PROTEIN, VACCINE COMPOSITION AGAINST LEISHMANIOSIS AND USES
BRBR1020160061210 2016-03-21

Publications (1)

Publication Number Publication Date
WO2017163171A1 true WO2017163171A1 (en) 2017-09-28

Family

ID=59914225

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2017/051610 WO2017163171A1 (en) 2016-03-21 2017-03-20 Chimeric protein, vaccine composition against leishmaniasis, and uses thereof

Country Status (2)

Country Link
BR (1) BR102016006121A2 (en)
WO (1) WO2017163171A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019159196A1 (en) * 2018-02-15 2019-08-22 National Centre For Cell Science A novel chimeric protein kinase c as an immunomodulator

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
MARTINS, V. T. ET AL.: "A Leishmania-specific hypothetical protein expressed in both promastigote and amastigote stages of Leishmania infantum employed for the serodiagnosis of, and as a vaccine candidate against, visceral leishmaniasis", PARASIT VECTORS., vol. 8, no. 363, July 2015 (2015-07-01) *
MARTINS, V. T. ET AL.: "Antigenicity and Protective Efficacy of a Leishmania Amastigote-specific Protein, Member of the Super- oxygenase Family, against Visceral Leishmaniasis", PLOS NEGL TROP DIS., vol. 7, no. 3, 2013, pages e2148 *
MARTINS, V. T. ET AL.: "Antigenicity, Immunogenicity and Protective Efficacy of Three Proteins Expressed in the Promastigote and Amastigote Stages of Leishmania infantum against Visceral Leishmaniasis", PLOS ONE, vol. 10, no. 9, 14 September 2016 (2016-09-14) *
MARTINS, V., T. ET AL.: "A recombinant chimeric protein composed by human and mice-specific CD 4 and CD 8 T cell epitopes protects against visceral leishmaniasis", PARASITE IMMUNOLOGY, vol. 12359, 16 September 2016 (2016-09-16), pages 1 - 14 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019159196A1 (en) * 2018-02-15 2019-08-22 National Centre For Cell Science A novel chimeric protein kinase c as an immunomodulator

Also Published As

Publication number Publication date
BR102016006121A2 (en) 2017-09-26

Similar Documents

Publication Publication Date Title
ES2670859T3 (en) Peptide sequences and compositions of Anopheles gambiae
Solioz et al. The protective capacities of histone H1 against experimental murine cutaneous leishmaniasis
Pacífico et al. Immunization with Schistosoma mansoni 22.6 kDa antigen induces partial protection against experimental infection in a recombinant protein form but not as DNA vaccine
ES2760004T3 (en) Leishmania vaccine using a salivary sand fly immunogen
BRPI0719078A2 (en) CANINE LEISHMANIA VACCINE
Ribeiro et al. Evaluation of a Leishmania hypothetical protein administered as DNA vaccine or recombinant protein against Leishmania infantum infection and its immunogenicity in humans
WO2017163171A1 (en) Chimeric protein, vaccine composition against leishmaniasis, and uses thereof
BRPI1015136B1 (en) SCHISOSOMOSIS VACCINE
ES2546301T3 (en) Composition containing Leishmania Lip2a
BR112015015539B1 (en) MONOVALENT AND MULTIVALENT VACCINES AGAINST LEISHMANIA
WO2018145182A1 (en) Hybrid peptide, set of hybrid peptides, composition, uses of the hybrid peptide, method for inducing an immune response and kits
BR102020021166A2 (en) Pharmaceutical and/or immunogenic compositions against visceral leishmaniasis and use
BR102018070807A2 (en) RECOMBINANT CHIMERIC PROTEIN, VACCINE AGAINST TEGUMENTARY AND VISCERAL LEISHMANIASIS AND USES
BR102020008111A2 (en) VACCINATION COMPOSITION AGAINST VISCERAL LEISHMANIASIS AND USE
BR102018016046A2 (en) VACCINE COMPOSITIONS AGAINST TEGUMENTARY AND VISCERAL LEISHMANIASIS AND USE
WO2023077206A1 (en) Genetically modified epitopes and multiepitope proteins, immunogenic compositions and use thereof
BR102014022027B1 (en) Immunobiological to control the leishmaniasis vector, obtaining processes and uses
BR102021010469A2 (en) RECOMBINANT CHIMERIC PROTEIN, ITS USE AND COMPOSITION
BR112016012097A2 (en) vaccine composition for the prevention and / or treatment of leishmaniasis, immunogenic peptides and obtaining process
Al-Idreesi et al. Immune responses to Eimeria tenella sporozoite protein as vaccine to broiler against coccidiosis
BR112020024211A2 (en) multiepitopic peptide compounds and vaccines against leishmaniasis
WO2012061910A2 (en) Main immunoprotective domains and epitopes of nucleoside hydrolase from leishmania (l) donovani for use in vaccination, immunotherapy and diagnosis
BRPI1015788B1 (en) main immunoprotective domains and epitopes of leishmania (l.) donovani nucleoside hydrolase for use in vaccination
JP2002533474A (en) El. Chimeric genes encoding antigenic determinants of four proteins of L. infantum
AU2013200258A1 (en) Peptide sequences and compositions

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17769543

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17769543

Country of ref document: EP

Kind code of ref document: A1