Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Sci Rep ; 8(1): 1118, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348479

ABSTRACT

Vaccine development against Plasmodium vivax malaria lags behind that for Plasmodium falciparum. To narrow this gap, we administered recombinant antigens based on P. vivax circumsporozoite protein (CSP) to mice. We expressed in Pichia pastoris two chimeric proteins by merging the three central repeat regions of different CSP alleles (VK210, VK247, and P. vivax-like). The first construct (yPvCSP-AllFL) contained the fused repeat regions flanked by N- and C-terminal regions. The second construct (yPvCSP-AllCT) contained the fused repeat regions and the C-terminal domain, plus RI region. Mice were vaccinated with three doses of yPvCSP in adjuvants Poly (I:C) or Montanide ISA720. We also used replication-defective adenovirus vectors expressing CSP of human serotype 5 (AdHu5) and chimpanzee serotype 68 (AdC68) for priming mice which were subsequently boosted twice with yPvCSP proteins in Poly (I:C) adjuvant. Regardless of the regime used, immunized mice generated high IgG titres specific to all CSP alleles. After challenge with P. berghei ANKA transgenic parasites expressing Pb/PvVK210 or Pb/PvVK247 sporozoites, significant time delays for parasitemia were observed in all vaccinated mice. These vaccine formulations should be clinically tried for their potential as protective universal vaccine against P. vivax malaria.


Subject(s)
Malaria Vaccines/immunology , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Recombinant Proteins/immunology , Adenoviridae/genetics , Amino Acid Sequence , Animals , Antibodies, Protozoan/immunology , Antibody Affinity/immunology , Disease Models, Animal , Female , Genetic Vectors/administration & dosage , Genetic Vectors/chemistry , Immunization , Immunogenicity, Vaccine , Immunoglobulin G/blood , Immunoglobulin G/immunology , Malaria Vaccines/genetics , Malaria, Vivax/mortality , Mice , Plasmodium vivax/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
2.
Front Immunol ; 8: 1275, 2017.
Article in English | MEDLINE | ID: mdl-29075260

ABSTRACT

Plasmodium vivax is the most common species that cause malaria outside of the African continent. The development of an efficacious vaccine would contribute greatly to control malaria. Recently, using bacterial and adenoviral recombinant proteins based on the P. vivax circumsporozoite protein (CSP), we demonstrated the possibility of eliciting strong antibody-mediated immune responses to each of the three allelic forms of P. vivax CSP (PvCSP). In the present study, recombinant proteins representing the PvCSP alleles (VK210, VK247, and P. vivax-like), as well as a hybrid polypeptide, named PvCSP-All epitopes, were generated. This hybrid containing the conserved C-terminal of the PvCSP and the three variant repeat domains in tandem were successfully produced in the yeast Pichia pastoris. After purification and biochemical characterization, they were used for the experimental immunization of C57BL/6 mice in a vaccine formulation containing the adjuvant Poly(I:C). Immunization with a recombinant protein expressing all three different allelic forms in fusion elicited high IgG antibody titers reacting with all three different allelic variants of PvCSP. The antibodies targeted both the C-terminal and repeat domains of PvCSP and recognized the native protein on the surface of P. vivax sporozoites. More importantly, mice that received the vaccine formulation were protected after challenge with chimeric Plasmodium berghei sporozoites expressing CSP repeats of P. vivax sporozoites (Pb/PvVK210). Our results suggest that it is possible to elicit protective immunity against one of the most common PvCSP alleles using soluble recombinant proteins expressed by P. pastoris. These recombinant proteins are promising candidates for clinical trials aiming to develop a multiallele vaccine against P. vivax malaria.

3.
Vaccine ; 34(38): 4501-4506, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27502569

ABSTRACT

A number of studies have shown that CD8+ T cells mediate protective anti-malaria immunity in a mouse model. However, whether human CD8+ T cells play a role in protection against malaria remains unknown. We recently established human immune system (HIS) mice harboring functional human CD8+ T cells (HIS-CD8 mice) by transduction with HLA-A∗0201 and certain human cytokines using recombinant adeno-associated virus-based gene transfer technologies. These HIS-CD8 mice mount a potent, antigen-specific HLA-A∗0201-restricted human CD8+ T-cell response upon immunization with a recombinant adenovirus expressing a human malaria antigen, the Plasmodium falciparum circumsporozoite protein (PfCSP), termed AdPfCSP. In the present study, we challenged AdPfCSP-immunized HIS-CD8 mice with transgenic Plasmodium berghei sporozoites expressing full-length PfCSP and found that AdPfCSP-immunized (but not naïve) mice were protected against subsequent malaria challenge. The level of the HLA-A∗0201-restricted, PfCSP-specific human CD8+ T-cell response was closely correlated with the level of malaria protection. Furthermore, depletion of human CD8+ T cells from AdPfCSP-immunized HIS-CD8 mice almost completely abolished the anti-malaria immune response. Taken together, our data show that human CD8+ T cells mediate protective anti-malaria immunity in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Animals , Dependovirus , Genetic Vectors , HLA-A2 Antigen/genetics , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Plasmodium falciparum , Protozoan Proteins/immunology , Sporozoites
4.
J Immunol Methods ; 427: 42-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26410104

ABSTRACT

In this study, we developed human immune system (HIS) mice that possess functional human CD4+ T cells and B cells, named HIS-CD4/B mice. HIS-CD4/B mice were generated by first introducing HLA class II genes, including DR1 and DR4, along with genes encoding various human cytokines and human B cell activation factor (BAFF) to NSG mice by adeno-associated virus serotype 9 (AAV9) vectors, followed by engrafting human hematopoietic stem cells (HSCs). HIS-CD4/B mice, in which the reconstitution of human CD4+ T and B cells resembles to that of humans, produced a significant level of human IgG against Plasmodium falciparum circumsporozoite (PfCS) protein upon immunization. CD4+ T cells in HIS-CD4/B mice, which possess central and effector memory phenotypes like those in humans, are functional, since PfCS protein-specific human CD4+ T cells secreting IFN-γ and IL-2 were detected in immunized HIS-CD4/B mice. Lastly, PfCS protein-immunized HIS-CD4/B mice were protected from in vivo challenge with transgenic P. berghei sporozoites expressing the PfCS protein. The immune sera collected from protected HIS-CD4/B mice reacted against transgenic P. berghei sporozoites expressing the PfCS protein and also inhibited the parasite invasion into hepatocytes in vitro. Taken together, these studies show that our HIS-CD4/B mice could mount protective human anti-malaria immunity, consisting of human IgG and human CD4+ T cell responses both specific for a human malaria antigen.


Subject(s)
Disease Models, Animal , Immunity, Humoral/immunology , Malaria, Falciparum/immunology , Mice, Transgenic/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Heterografts , Histocompatibility Antigens Class II , Humans , Malaria Vaccines , Mice , Protozoan Proteins/immunology
5.
Infect Immun ; 82(2): 793-807, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24478093

ABSTRACT

Plasmodium vivax is the most widespread and the second most prevalent malaria-causing species in the world. Current measures used to control the transmission of this disease would benefit from the development of an efficacious vaccine. In the case of the deadly parasite P. falciparum, the recombinant RTS,S vaccine containing the circumsporozoite antigen (CSP) consistently protects 30 to 50% of human volunteers against infection and is undergoing phase III clinical trials in Africa with similar efficacy. These findings encouraged us to develop a P. vivax vaccine containing the three circulating allelic forms of P. vivax CSP. Toward this goal, we generated three recombinant bacterial proteins representing the CSP alleles, as well as a hybrid polypeptide called PvCSP-All-CSP-epitopes. This hybrid contains the conserved N and C termini of P. vivax CSP and the three variant repeat domains in tandem. We also generated simian and human recombinant replication-defective adenovirus vectors expressing PvCSP-All-CSP-epitopes. Mice immunized with the mixture of recombinant proteins in a formulation containing the adjuvant poly(I·C) developed high and long-lasting serum IgG titers comparable to those elicited by proteins emulsified in complete Freund's adjuvant. Antibody titers were similar in mice immunized with homologous (protein-protein) and heterologous (adenovirus-protein) vaccine regimens. The antibodies recognized the three allelic forms of CSP, reacted to the repeated and nonrepeated regions of CSP, and recognized sporozoites expressing the alleles VK210 and VK247. The vaccine formulations described in this work should be useful for the further development of an anti-P. vivax vaccine.


Subject(s)
Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/blood , Female , Immunoglobulin G/blood , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria, Vivax/immunology , Mice , Mice, Inbred C57BL , Plasmodium vivax/genetics , Poly I-C/administration & dosage , Protozoan Proteins/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
6.
J Immunol Methods ; 377(1-2): 47-52, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22306356

ABSTRACT

Plasmodium sporozoites are deposited in the skin of the mammalian host by Anopheles mosquitoes. To continue the life cycle, the sporozoites have to invade the host's hepatocytes, where they transform into exoerythrocytic forms (EEFs) inside a parasitophorous vacuole. During their route from the skin to the liver, the parasites traverse the capillary epithelium in the dermis to enter the blood circulation, and cross the endothelium of liver sinusoids to enter the parenchyma. Cell traversal by sporozoites is usually measured by quantifying dyes that enter or are released from cells during incubation with salivary gland sporozoites. These methods do not distinguish cell traversal from cell wounding. Here we validate an assay that quantifies cell traversal of sporozoites through monolayers of MDCK cells that form tight junctions. We compared cell traversal of wt sporozoites and of parasites lacking the Type I membrane protein TLP (TRAP-like protein) previously implicated in cell traversal. We provide direct evidence that TLP ko sporozoites are defective in cell traversal and that they are retained inside the MDCK cytoplasm. We then used the MDCK assay to study the effect of a monoclonal antibody (3D11) to the circumsporozoite protein (CSP) on the parasite's cell traversal. We show that 3D11 inhibits cell traversal at nanomolar concentrations. We conclude that antibodies elicited by CSP-based vaccines are likely to inhibit the migration of sporozoites from the skin to the liver.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Animals , Cell Line , Dogs , Electric Impedance , Intracellular Signaling Peptides and Proteins/immunology , Tight Junctions/immunology
7.
Mem Inst Oswaldo Cruz ; 106 Suppl 1: 167-71, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21881771

ABSTRACT

Recently, we described the improved immunogenicity of new malaria vaccine candidates based on the expression of fusion proteins containing immunodominant epitopes of merozoites and Salmonella enterica serovar Typhimurium flagellin (FliC) protein as an innate immune agonist. Here, we tested whether a similar strategy, based on an immunodominant B-cell epitope from malaria sporozoites, could also generate immunogenic fusion polypeptides. A recombinant His6-tagged FliC protein containing the C-terminal repeat regions of the VK210 variant of Plasmodium vivax circumsporozoite (CS) protein was constructed. This recombinant protein was successfully expressed in Escherichia coli as soluble protein and was purified by affinity to Ni-agarose beads followed by ion exchange chromatography. A monoclonal antibody specific for the CS protein of P. vivax sporozoites (VK210) was able to recognise the purified protein. C57BL/6 mice subcutaneously immunised with the recombinant fusion protein in the absence of any conventional adjuvant developed protein-specific systemic antibody responses. However, in mice genetically deficient in expression of TLR5, this immune response was extremely low. These results extend our previous observations concerning the immunogenicity of these recombinant fusion proteins and provide evidence that the main mechanism responsible for this immune activation involves interactions with TLR5, which has not previously been demonstrated for any recombinant FliC fusion protein.


Subject(s)
Flagellin/immunology , Immunodominant Epitopes/immunology , Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Plasmodium falciparum/immunology , Recombinant Fusion Proteins/immunology , Salmonella typhimurium/immunology , Animals , Antibodies, Protozoan/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , Escherichia coli Proteins/immunology , Flagellin/metabolism , Immunodominant Epitopes/metabolism , Malaria Vaccines/metabolism , Malaria, Vivax/immunology , Mice , Mice, Inbred C57BL , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Salmonella typhimurium/metabolism , Toll-Like Receptor 5/immunology
8.
Vaccine ; 29(43): 7335-42, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21807053

ABSTRACT

Immunization of BALB/c mice with irradiated sporozoites (IrSp) of Plasmodium yoelii can lead to sterile immunity. The circumsporozoite protein (CSP) plays a dominant role in protection. Nevertheless after hyper-immunization with IrSp, complete protection is obtained in CSP-transgenic BALB/c mice that are T-cell tolerant to the CSP and cannot produce antibodies [CSP-Tg/JhT(-/-)]. This protection is mediated exclusively by CD8(+) T cells [1]. To identify the non-CSP protective T cell antigens, we studied the properties of 34 P. yoelii sporozoite antigens that are predicted to be secreted and to contain strong Kd-restricted CD8(+) T cell epitopes. The synthetic peptides corresponding to the epitopes were used to screen for the presence of peptide-specific CD8(+) T cells secreting interferon-γ (IFN-γ) in splenocytes from CSP-Tg/JhT(-/-) BALB/c mice hyper immunized with IrSp. However, the numbers of IFN-γ-secreting splenocytes specific for the non-CSP antigen-derived peptides were 20-100 times lower than those specific for the CSP-specific peptide. When mice were immunized with recombinant adenoviruses expressing selected non-CSP antigens, the animals were not protected against challenge with P. yoelii sporozoites although large numbers of CD8(+) specific T cells were generated.


Subject(s)
Antigens, Protozoan/immunology , Epitopes, T-Lymphocyte/immunology , Malaria Vaccines/immunology , Plasmodium yoelii/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Immunization , Interferon-gamma , Malaria/immunology , Malaria/prevention & control , Mice , Mice, Inbred BALB C , Mice, Transgenic , Sporozoites/radiation effects
9.
Mem. Inst. Oswaldo Cruz ; 106(supl.1): 167-171, Aug. 2011. ilus, graf
Article in English | LILACS | ID: lil-597258

ABSTRACT

Recently, we described the improved immunogenicity of new malaria vaccine candidates based on the expression of fusion proteins containing immunodominant epitopes of merozoites and Salmonella enterica serovar Typhimurium flagellin (FliC) protein as an innate immune agonist. Here, we tested whether a similar strategy, based on an immunodominant B-cell epitope from malaria sporozoites, could also generate immunogenic fusion polypeptides. A recombinant His6-tagged FliC protein containing the C-terminal repeat regions of the VK210 variant of Plasmodium vivax circumsporozoite (CS) protein was constructed. This recombinant protein was successfully expressed in Escherichia coli as soluble protein and was purified by affinity to Ni-agarose beads followed by ion exchange chromatography. A monoclonal antibody specific for the CS protein of P. vivax sporozoites (VK210) was able to recognise the purified protein. C57BL/6 mice subcutaneously immunised with the recombinant fusion protein in the absence of any conventional adjuvant developed protein-specific systemic antibody responses. However, in mice genetically deficient in expression of TLR5, this immune response was extremely low. These results extend our previous observations concerning the immunogenicity of these recombinant fusion proteins and provide evidence that the main mechanism responsible for this immune activation involves interactions with TLR5, which has not previously been demonstrated for any recombinant FliC fusion protein.


Subject(s)
Animals , Mice , Flagellin/immunology , Immunodominant Epitopes/immunology , Malaria Vaccines/immunology , Malaria, Vivax , Plasmodium falciparum/immunology , Recombinant Fusion Proteins/immunology , Salmonella typhimurium/immunology , Antibodies, Protozoan/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte , Escherichia coli Proteins/immunology , Flagellin , Immunodominant Epitopes , Malaria Vaccines , Malaria, Vivax/immunology , Protozoan Proteins/immunology , Protozoan Proteins , Recombinant Fusion Proteins , Salmonella typhimurium , /immunology
10.
Hist Cienc Saude Manguinhos ; 18(2): 559-64, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21779699

ABSTRACT

Since the 1960s, the scientist Ruth Nussenzweig, C.V. Starr Professor at New York University School of Medicine, has been working to develop an antimalarial vaccine. In her testimony, she traces some of the stages through which her research has passed. At the beginning of her studies, most scientists held that it would be impossible to develop such a vaccine. However, a different opinion had been expressed in a paper on avian malaria written some forty years earlier by a British researcher and his collaborators from India. The immunization principle developed by this group was irradiation of sporozoites in order to deactivate the parasite that causes malaria. Ruth Nussenzweig revived and expanded upon this line of research, which now underpins her efforts to devise an antimalarial vaccine for human use.

11.
Hist. ciênc. saúde-Manguinhos ; 18(2): 559-564, abr.-jun. 2011. ilus
Article in English | LILACS | ID: lil-593159

ABSTRACT

Since the 1960s, the scientist Ruth Nussenzweig, C.V. Starr Professor atNew York University School of Medicine, has been working to developan antimalarial vaccine. In her testimony, she traces some of the stagesthrough which her research has passed. At the beginning of herstudies, most scientists held that it would be impossible to developsuch a vaccine. However, a different opinion had been expressed in apaper on avian malaria written some forty years earlier by a Britishresearcher and his collaborators from India. The immunizationprinciple developed by this group was irradiation of sporozoites inorder to deactivate the parasite that causes malaria. Ruth Nussenzweigrevived and expanded upon this line of research, which nowunderpins her efforts to devise an antimalarial vaccine for human use


Subject(s)
Allergy and Immunology , Malaria/prevention & control , Parasitology , Vaccines
12.
J Exp Med ; 207(7): 1465-74, 2010 Jul 05.
Article in English | MEDLINE | ID: mdl-20584882

ABSTRACT

Sporozoites, the invasive form of malaria parasites transmitted by mosquitoes, are quiescent while in the insect salivary glands. Sporozoites only differentiate inside of the hepatocytes of the mammalian host. We show that sporozoite latency is an active process controlled by a eukaryotic initiation factor-2alpha (eIF2alpha) kinase (IK2) and a phosphatase. IK2 activity is dominant in salivary gland sporozoites, leading to an inhibition of translation and accumulation of stalled mRNAs into granules. When sporozoites are injected into the mammalian host, an eIF2alpha phosphatase removes the PO4 from eIF2alpha-P, and the repression of translation is alleviated to permit their transformation into liver stages. In IK2 knockout sporozoites, eIF2alpha is not phosphorylated and the parasites transform prematurely into liver stages and lose their infectivity. Thus, to complete their life cycle, Plasmodium sporozoites exploit the mechanism that regulates stress responses in eukaryotic cells.


Subject(s)
Culicidae/parasitology , Plasmodium berghei/enzymology , Salivary Glands/parasitology , Sporozoites/enzymology , eIF-2 Kinase/metabolism , Animals , Cell Line , Cytoplasmic Granules/metabolism , Gene Expression Regulation , Gene Targeting , Life Cycle Stages , Liver/metabolism , Liver/parasitology , Mice , Mice, Inbred C57BL , Phenotype , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Plasmodium berghei/cytology , Plasmodium berghei/pathogenicity , Plasmodium berghei/ultrastructure , Protein Biosynthesis , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Salivary Glands/cytology , Salivary Glands/ultrastructure , Sporozoites/cytology , Sporozoites/ultrastructure
13.
Vaccine ; 28(29): 4644-52, 2010 Jun 23.
Article in English | MEDLINE | ID: mdl-20451637

ABSTRACT

The live-attenuated yellow fever vaccine (YF17D) is one of the safest and most effective vaccines available today. Here, YF17D was genetically altered to express the circumsporozoite protein (CSP) from the murine malarial parasite Plasmodium yoelii. Reconstituted recombinant virus was viable and exhibited robust CSP expression. Immunization of naïve mice resulted in extensive proliferation of adoptively transferred CSP-specific transgenic CD8(+) T-cells. A single immunization of naïve mice with recombinant YF17D resulted in robust production of IFN-gamma by CD8(+) T-cells and IFN-gamma and IL-2 by CD4(+) T-cells. A prime-boost regimen consisting of recombinant virus followed by a low-dose of irradiated sporozoites conferred protection against challenge with P. yoelii. Taken together, these results show that recombinant YF17D can efficiently express CSP in culture, and prime a protective immune response in vivo.


Subject(s)
Malaria Vaccines/immunology , Malaria/prevention & control , Protozoan Proteins/immunology , Yellow Fever Vaccine/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Immunity, Cellular , Immunization, Secondary , Interferon-gamma/immunology , Interleukin-2/immunology , Malaria/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Plasmids , Plasmodium yoelii/immunology , Vaccines, Attenuated/immunology
14.
Proc Natl Acad Sci U S A ; 107(3): 1172-7, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20080619

ABSTRACT

There is yet no licensed vaccine against malaria, a serious human disease affecting mostly children, with an annual death rate of about one million. Plasmodia, the malaria-causing parasites, have two obligatory hosts: mammals or birds, in which they multiply asexually, and mosquitoes with sexual multiplication. The most common and serious type of malaria is caused by Plasmodium falciparum. The circumsporozoite protein (CSP), a major surface antigen of sporozoites, is a protective antigen. A unique feature of P. falciparum CSP is its large central domain composed of over 30 tetrapeptide repeats of Asn-Ala-Asn-Pro (NANP). Several NANP peptide-protein conjugates were tested clinically but elicited a low level of CSP antibodies for a short duration. To provide a CSP-based candidate vaccine, we investigated recombinant CSP and NANP conjugates of various peptide lengths, with different N-terminal amino acids, bound at different ratios to various carrier proteins. Injected into mice, CSP alone and CSP or NANP conjugates induced antibodies with booster responses and were positive by the sporozoite immunofluorescent assay. The use of the mosquito stage P. falciparum ookinete surface protein, Pfs25, cross-linked onto itself as a carrier for NANP, induced in mice high levels of uniquely long-lasting antibodies to both vaccine components with secondary biological activities, that will provide immunity to liver infection by sporozoites and block transmission by mosquitoes.


Subject(s)
Malaria Vaccines/immunology , Peptides/chemistry , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Animals , Antibodies, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Female , Fluorescent Antibody Technique , Gas Chromatography-Mass Spectrometry , Malaria Vaccines/chemistry , Mice , Peptides/immunology , Protozoan Proteins/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , T-Lymphocytes/immunology
15.
Hum Vaccin ; 6(1): 90-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19806009

ABSTRACT

The RTS,S/AS01(E) malaria vaccine candidate has recently entered Phase 3 testing. Reaching this important milestone is the culmination of more than 20 years of research and development by GlaxoSmithKline and partners and collaborators. The vaccine has been developed to protect young children and infants living in Sub-Saharan Africa against clinical and severe disease caused by Plasmodium falciparum infection. Over the past 9 years, RTS,S/AS has been evaluated in multiple Phase 2 studies. The vaccine was shown to have a favorable safety profile and to be well tolerated in all age groups in which it was tested, including the intended target population of infants and young children in Sub-Saharan Africa. Data obtained so far suggest that RTS,S/AS can be co-administered with other vaccines included in the routine Expanded Program of Immunization (EPI). In Phase 2 testing, the vaccine candidate was shown to confer significant protection against P. falciparum infection and clinical disease, including severe malaria. Furthermore, a trend towards an indirect beneficial effect of the vaccine on non-malarial morbidities has been observed in several trials. In this paper, we will describe the genesis of the RTS,S/AS concept, including the rationale for selecting the circumsporozoite protein (CSP) as the target antigen. Early development history of the vaccine will be briefly described. We will present the most salient results from recent Phase 2 studies conducted in the target pediatric population, which have led to the decision to progress RTS,S/AS to Phase 3 testing. If the Phase 3 results confirm the observations made during Phase 2 testing, the RTS,S/AS vaccine, when broadly implemented and judiciously integrated with other malaria-prevention measures, would have a major public-health impact in Sub-Saharan Africa.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Africa South of the Sahara/epidemiology , Child, Preschool , Clinical Trials as Topic , Humans , Infant , Malaria Vaccines/adverse effects , Protozoan Proteins/immunology
16.
Nature ; 444(7121): 937-40, 2006 Dec 14.
Article in English | MEDLINE | ID: mdl-17151604

ABSTRACT

Malaria infection starts when mosquitoes inject sporozoites into the skin. The parasites enter the blood stream and make their way to the liver where they develop into the exo-erythrocytic forms (EEFs). Immunization with irradiated sporozoites (IrSp) leads to robust protection against malaria infection in rodents, monkeys and humans by eliciting antibodies to circumsporozoite protein (CS) that inhibit sporozoite infectivity, and T cells that destroy the EEFs. To study the role of non-CS antigens in protection, we produced CS transgenic mice that were tolerant to CS T-cell epitopes. Here we show that in the absence of T-cell-dependent immune responses to CS, protection induced by immunization with two doses of IrSp was greatly reduced. Thus, although hundreds of other Plasmodium genes are expressed in sporozoites and EEFs, CS is a dominant protective antigen. Nevertheless, sterile immunity could be obtained by immunization of CS transgenics with three doses of IrSp.


Subject(s)
Antigens, Protozoan/immunology , Immunodominant Epitopes/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Sporozoites/radiation effects , Animals , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Sporozoites/chemistry , T-Lymphocytes, Cytotoxic/immunology
17.
J Exp Med ; 203(3): 599-606, 2006 Mar 20.
Article in English | MEDLINE | ID: mdl-16505139

ABSTRACT

Resistance to several prevalent infectious diseases requires both cellular and humoral immune responses. T cell immunity is initiated by mature dendritic cells (DCs) in lymphoid organs, whereas humoral responses to most antigens require further collaboration between primed, antigen-specific helper T cells and naive or memory B cells. To determine whether antigens delivered to DCs in lymphoid organs induce T cell help for antibody responses, we targeted a carrier protein, ovalbumin (OVA), to DCs in the presence of a maturation stimulus and assayed for antibodies to a hapten, (4-hydroxy-3-nitrophenyl) acetyl (NP), after boosting with OVA-NP. A single DC-targeted immunization elicited long-lived T cell helper responses to the carrier protein, leading to large numbers of antibody-secreting cells and high titers of high-affinity antihapten immunoglobulin Gs. Small doses of DC-targeted OVA induced higher titers and a broader spectrum of anti-NP antibody isotypes than large doses of OVA in alum adjuvant. Similar results were obtained when the circumsporozoite protein of Plasmodium yoelii was delivered to DCs. We conclude that antigen targeting to DCs combined with a maturation stimulus produces broad-based and long-lived T cell help for humoral immune responses.


Subject(s)
Antibody Formation/immunology , Antigen Presentation/immunology , B-Lymphocytes/immunology , Dendritic Cells/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Animals , Antibody Formation/drug effects , Antigen Presentation/drug effects , Chickens , Haptens/immunology , Humans , Immunization/methods , Immunoglobulin G/immunology , Immunologic Memory/drug effects , Immunologic Memory/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Plasmodium yoelii/immunology , Protozoan Proteins/immunology
18.
Infect Immun ; 73(6): 3587-97, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15908388

ABSTRACT

Highly purified subunit vaccines require potent adjuvants in order to elicit optimal immune responses. In a previous phase I trial, an alum formulation of ICC-1132, a malaria vaccine candidate comprising hepatitis B core (HBc) virus-like particle containing Plasmodium falciparum circumsporozoite (CS) protein epitopes, was shown to elicit Plasmodium falciparum-specific antibody and cellular responses. The present study was designed as a single-blind, escalating-dose phase I trial to evaluate the safety and immunogenicity of single intramuscular doses of ICC-1132 formulated in the more potent water-in-oil adjuvant Montanide ISA 720 (ICC-1132/ISA 720). The vaccine was safe and well tolerated, with transient injection site pain as the most frequent complaint. All vaccinees that received either 20 mug or 50 mug of ICC-1132/ISA 720 developed antiimmunogen and anti-HBc antibodies. The majority of volunteers in these two groups developed sporozoite-specific antibodies, predominantly of opsonizing immunoglobulin G subtypes. Peak titers and persistence of parasite-specific antibody following a single injection of the ISA 720 formulated vaccine were comparable to those obtained following two to three immunizations with alum-adsorbed ICC-1132. Peripheral blood mononuclear cells of ICC-1132/ISA 720 vaccinees proliferated and released cytokines (interleukin 2 and gamma interferon) when stimulated with recombinant P. falciparum CS protein, and CS-specific CD4(+) T-cell lines were established from volunteers with high levels of antibodies to the repeat region. The promising results obtained with a single dose of ICC-1132 formulated in Montanide ISA 720 encourage further clinical development of this malaria vaccine candidate.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Hepatitis B Core Antigens/administration & dosage , Malaria Vaccines/administration & dosage , Mannitol/analogs & derivatives , Mannitol/administration & dosage , Oleic Acids/administration & dosage , Plasmodium falciparum/immunology , Adult , Amino Acid Sequence , Animals , Antibodies, Protozoan/blood , Antibody Specificity , CD4-Positive T-Lymphocytes/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin G/classification , Interferon-gamma/biosynthesis , Lymphocyte Activation , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Molecular Sequence Data , Single-Blind Method
19.
J Exp Med ; 201(2): 201-9, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15657290

ABSTRACT

The yellow fever vaccine 17D (17D) is safe, and after a single immunizing dose, elicits long-lasting, perhaps lifelong protective immunity. One of the major challenges facing delivery of human vaccines in underdeveloped countries is the need for multiple injections to achieve full efficacy. To examine 17D as a vector for microbial T cell epitopes, we inserted the H-2K(d)-restricted CTL epitope of the circumsporozoite protein (CS) of Plasmodium yoelii between 17D nonstructural proteins NS2B and NS3. The recombinant virus, 17D-Py, was replication competent and stable in vitro and in vivo. A single subcutaneous injection of 10(5) PFU diminished the parasite burden in the liver by approximately 70%. The high level of protection lasted between 4 and 8 wk after immunization, but a significant effect was documented even 24 wk afterwards. Thus, the immunogenicity of a foreign T cell epitope inserted into 17D mimics some of the remarkable properties of the human vaccine. Priming with 17D-Py followed by boosting with irradiated sporozoites conferred sterile immunity to 90% of the mice. This finding indicates that the immune response of vaccine-primed individuals living in endemic areas could be sustained and magnified by the bite of infected mosquitoes.


Subject(s)
Epitopes/immunology , Malaria/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Yellow Fever Vaccine/immunology , Animals , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Malaria/immunology , Mice , Plasmodium yoelii/immunology , Yellow Fever Vaccine/genetics , Yellow fever virus/genetics , Yellow fever virus/immunology
20.
J Virol ; 77(21): 11859-66, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14557672

ABSTRACT

We immunized mice with an attenuated (cold-adapted) influenza virus followed by an attenuated vaccinia virus (modified vaccinia virus Ankara), both expressing a CD8(+)-T-cell epitope derived from malaria sporozoites. This vaccination regimen elicited high levels of protection against malaria. This is the first time that the vaccine efficacy of a recombinant cold-adapted influenza virus vector expressing a foreign antigen has been evaluated.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Malaria Vaccines/immunology , Malaria/prevention & control , Orthomyxoviridae/immunology , Plasmodium yoelii/immunology , Protozoan Proteins/immunology , Vaccinia virus/immunology , Adaptation, Physiological , Animals , Antibodies, Protozoan/blood , Cold Temperature , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Female , Genetic Vectors , Humans , Immunization , Immunization, Secondary , Malaria/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mice , Mice, Inbred BALB C , Orthomyxoviridae/genetics , Plasmodium yoelii/genetics , Protozoan Proteins/genetics , Recombination, Genetic , Vaccinia virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...