ABSTRACT
Malaria represents a serious public health problem, presenting with high rates of incidence, morbidity and mortality in tropical and subtropical regions of the world. According to the World Health Organization, in 2018 there were 228 million cases and 405 thousand deaths caused by this disease in the world, affecting mainly children and pregnant women in Africa. Despite the programs carried out to control this disease, drug resistance and invertebrate vector resistance to insecticides have generated difficulties. An efficient vaccine against malaria would be a strategy with a high impact on the eradication and control of this disease. Researches aimed at developing vaccines have focused on antigens of high importance for the survival of the parasite such as the Circumsporozoite Surface Protein, involved in the pre-erythrocytic cycle during parasites invasion in hepatocytes. Currently, RTS'S is the most promising vaccine for malaria and was constructed using CSP; its performance was evaluated using two types of adjuvants: AS01 and AS02. The purpose of this review was to provide a bibliographic survey of historical researches that led to the development of RTS'S and its performance analysis over the decade. The search for new adjuvants to be associated with this antigen seems to be a way to obtain higher percentages of protection for a future malaria vaccine.
Subject(s)
Malaria Vaccines/therapeutic use , Malaria/prevention & control , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins , Humans , Malaria/parasitology , Malaria Vaccines/administration & dosage , Membrane ProteinsABSTRACT
Worldwide strategies between 2010 and 2017 aimed at controlling malarial parasites (mainly Plasmodium falciparum) led to a reduction of just 18% regarding disease incidence rates. Many biologically-derived anti-malarial vaccine candidates have been developed to date; this has involved using many experimental animals, an immense amount of work and the investment of millions of dollars. This review provides an overview of the current state and the main results of clinical trials for sporozoite-targeting vaccines (i.e. the parasite stage infecting the liver) carried out by research groups in areas having variable malaria transmission rates. However, none has led to promising results regarding the effective control of the disease, thereby making it necessary to complement such efforts at finding/introducing new vaccine candidates by adopting a multi-epitope, multi-stage approach, based on minimal subunits of the main sporozoite proteins involved in the invasion of the liver.
Subject(s)
Malaria Vaccines , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Animals , Anopheles/parasitology , Erythrocytes/parasitology , Humans , Liver/parasitology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/transmission , Mosquito Vectors/parasitology , Plasmodium falciparum/growth & development , Sporozoites/immunology , Sporozoites/radiation effects , Vaccines, Attenuated , Vaccines, Subunit , Vaccines, SyntheticABSTRACT
Introduction: Numerous challenges have hampered developing an anti-malarial vaccine against the most widespread malarial parasite worldwide: Plasmodium vivax. Despite the progress achieved in studying proteins in short-term in vitro culture or in experimental models, there is still no clear method for defining which antigens or their regions should be prioritized for including them in a vaccine.Areas covered: The methods used by research groups so far which have focused on the functional analysis of P. vivax blood stage antigens have been reviewed here. A logical strategy orientated toward resolving two of the most commonly occurring problems in designing vaccines against this species has thus been proposed (i.e. the search for candidates and evaluating/ascertaining their functional role in the invasion of such molecules).Expert commentary: Advances in knowledge regarding P. vivax biology have been extremely slow. Only two key receptor-ligand interactions concerning merozoite entry to reticulocytes have been reported during the last 20 years: PvDBP1-DARC and PvRBP2b-CD71. Despite increasing knowledge about the parasite's intimate preference for its host cells, it has yet to be determined which regions of the merozoite molecules characterized to date meet the requirement of inducing protective immune responses effectively blocking heterologous parasite entry to human cells.
Subject(s)
Malaria Vaccines/administration & dosage , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Animals , Antigens, Protozoan/immunology , Humans , Malaria Vaccines/immunology , Malaria, Vivax/immunologyABSTRACT
Plasmodium vivax is the most widely distributed malaria species and the most prevalent species of malaria in America and Asia. Vaccine development against P. vivax is considered a priority in the global program for the eradication of malaria. Earlier studies have characterized the Apical Membrane Antigen 1 (AMA-1) ectodomain and the C-terminal region (19kDa) of the Merozoite Surface Protein 1 (MSP-1) of P. vivax as immunodominant antigens. Based on this characterization, we designed a chimeric recombinant protein containing both merozoite immunodominant domains (PvAMA166-MSP119). The recombinant PvAMA166-MSP119 was successfully expressed in Pichia pastoris and used to immunize two different mouse strains (BALB/c and C57BL/6) in the presence of the Poly (I:C) as an adjuvant. Immunization with the chimeric protein induced high antibody titers against both proteins in both strains of mice as detected by ELISA. Antisera also recognized the native proteins expressed on the merozoites of mature P. vivax schizonts. Moreover, this antigen was able to induce IFN-gamma-secreting cells in C57BL/6 mice. These findings indicate that this novel yeast recombinant protein containing PvAMA166 and PvMSP119 is advantageous, because of improved antibody titers and cellular immune response. Therefore, this formulation should be further developed for pre-clinical trials in non-human primates as a potential candidate for a P. vivax vaccine.
Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Membrane Proteins/immunology , Merozoite Surface Protein 1/immunology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Recombinant Fusion Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Membrane Proteins/genetics , Merozoite Surface Protein 1/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Pichia/genetics , Poly I-C/administration & dosage , Protozoan Proteins/genetics , Recombinant Fusion Proteins/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunologyABSTRACT
BACKGROUND: Immunizing human volunteers by mosquito bite with radiation-attenuated Plasmodium falciparum sporozoites (RAS) results in high-level protection against infection. Only two volunteers have been similarly immunized with P. vivax (Pv) RAS, and both were protected. A phase 2 controlled clinical trial was conducted to assess the safety and protective efficacy of PvRAS immunization. METHODOLOGY/PRINCIPAL FINDINGS: A randomized, single-blinded trial was conducted. Duffy positive (Fy+; Pv susceptible) individuals were enrolled: 14 received bites from irradiated (150 ± 10 cGy) Pv-infected Anopheles mosquitoes (RAS) and 7 from non-irradiated non-infected mosquitoes (Ctl). An additional group of seven Fy- (Pv refractory) volunteers was immunized with bites from non-irradiated Pv-infected mosquitoes. A total of seven immunizations were carried out at mean intervals of nine weeks. Eight weeks after last immunization, a controlled human malaria infection (CHMI) with non-irradiated Pv-infected mosquitoes was performed. Nineteen volunteers completed seven immunizations (12 RAS, 2 Ctl, and 5 Fy-) and received a CHMI. Five of 12 (42%) RAS volunteers were protected (receiving a median of 434 infective bites) compared with 0/2 Ctl. None of the Fy- volunteers developed infection by the seventh immunization or after CHMI. All non-protected volunteers developed symptoms 8-13 days after CHMI with a mean pre-patent period of 12.8 days. No serious adverse events related to the immunizations were observed. Specific IgG1 anti-PvCS response was associated with protection. CONCLUSION: Immunization with PvRAS was safe, immunogenic, and induced sterile immunity in 42% of the Fy+ volunteers. Moreover, Fy- volunteers were refractory to Pv malaria. TRIAL REGISTRATION: Identifier: NCT01082341.
Subject(s)
Anopheles/parasitology , Immunization/methods , Insect Bites and Stings , Malaria Vaccines/immunology , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Adolescent , Adult , Animals , Antibodies, Protozoan/blood , Colombia , Duffy Blood-Group System , Female , Humans , Immunization/adverse effects , Immunoglobulin G/blood , Malaria Vaccines/administration & dosage , Malaria, Vivax/ethnology , Malaria, Vivax/parasitology , Male , Middle Aged , Plasmodium vivax/physiology , Plasmodium vivax/radiation effects , Single-Blind Method , Sporozoites/radiation effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Volunteers , Young AdultABSTRACT
Malaria remains a world-threatening disease largely because of the lack of a long-lasting and fully effective vaccine. MAEBL is a type 1 transmembrane molecule with a chimeric cysteine-rich ectodomain homologous to regions of the Duffy binding-like erythrocyte binding protein and apical membrane antigen 1 (AMA1) antigens. Although MAEBL does not appear to be essential for the survival of blood-stage forms, ectodomains M1 and M2, homologous to AMA1, seem to be involved in parasite attachment to erythrocytes, especially M2. MAEBL is necessary for sporozoite infection of mosquito salivary glands and is expressed in liver stages. Here, the Plasmodium yoelii MAEBL-M2 domain was expressed in a prokaryotic vector. C57BL/6J mice were immunized with doses of P. yoelii recombinant protein rPyM2-MAEBL. High levels of antibodies, with balanced IgG1 and IgG2c subclasses, were achieved. rPyM2-MAEBL antisera were capable of recognizing the native antigen. Anti-MAEBL antibodies recognized different MAEBL fragments expressed in CHO cells, showing stronger IgM and IgG responses to the M2 domain and repeat region, respectively. After a challenge with P. yoelii YM (lethal strain)-infected erythrocytes (IE), up to 90% of the immunized animals survived and a reduction of parasitemia was observed. Moreover, splenocytes harvested from immunized animals proliferated in a dose-dependent manner in the presence of rPyM2-MAEBL. Protection was highly dependent on CD4(+), but not CD8(+), T cells toward Th1. rPyM2-MAEBL antisera were also able to significantly inhibit parasite development, as observed in ex vivo P. yoelii erythrocyte invasion assays. Collectively, these findings support the use of MAEBL as a vaccine candidate and open perspectives to understand the mechanisms involved in protection.
Subject(s)
Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium yoelii/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/immunology , Erythrocytes/parasitology , Female , Humans , Immunization , Malaria/immunology , Malaria/mortality , Malaria/parasitology , Malaria Vaccines/administration & dosage , Malaria Vaccines/chemistry , Malaria Vaccines/genetics , Male , Merozoites/chemistry , Merozoites/growth & development , Merozoites/immunology , Mice , Mice, Inbred C57BL , Plasmodium yoelii/chemistry , Plasmodium yoelii/genetics , Plasmodium yoelii/growth & development , Protein Structure, Tertiary , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Sporozoites/chemistry , Sporozoites/growth & development , Sporozoites/immunologyABSTRACT
BACKGROUND: Several studies have recently demonstrated that the immune responses against malaria is governed by different factors, including the genetic components of the host. The IL-4 gene appears to be a strong candidate factor because of its role in the regulation of the Th2 response. The present study investigated the role of IL-4 polymorphisms in the development of IgG antibodies against PvAMA-1 and the IL-4 levels in individuals infected with Plasmodium vivax in a malaria endemic area in the Brazilian Amazon. METHODS: The study sample included 83 patients who were diagnosed with P. vivax infection using thick smear and confirmed by nested-PCR. The IL-4 -590C>T and IL-4 -33C>T polymorphisms were genotyped by PCR-RFLP, and the intron 3 VNTR was genotyped by PCR. A standardised ELISA protocol was used to measure the total IgG against PvAMA-1. The cytokine/chemokine levels were measured using a Milliplex multiplex assay (Millipore). All of the subjects were genotyped with 48 ancestry informative markers to determine the proportions of African, European and Amerindian ancestry using STRUCTURE software. RESULTS: Of the 83 patients, 60 (73%) produced IgG antibodies against PvAMA-1. A significant decrease in the percentage of respondents was observed among the primo-infected individuals. No significant differences were observed in the frequencies of genotypes and haplotypes among individuals who were positive or negative for IgG antibodies against PvAMA-1. Furthermore, no significant correlation was observed between the IL-4 polymorphisms, antibody levels, IL-4 levels, and parasitemia. CONCLUSIONS: This study indicated that the polymorphisms identified in the IL-4 gene are not likely to play a role in the regulation of the antibody response against PvAMA-1 and IL-4 production in vivax malaria.
Subject(s)
Antigens, Protozoan/administration & dosage , Endemic Diseases , Interleukin-4/genetics , Malaria Vaccines/administration & dosage , Malaria, Vivax/genetics , Membrane Proteins/administration & dosage , Plasmodium vivax/immunology , Polymorphism, Genetic , Protozoan Proteins/administration & dosage , Adolescent , Adult , Aged , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Brazil/epidemiology , Female , Humans , Immunoglobulin G/immunology , Interleukin-4/immunology , Malaria Vaccines/immunology , Malaria, Vivax/epidemiology , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Male , Membrane Proteins/immunology , Middle Aged , Protozoan Proteins/immunology , Th2 Cells/immunologyABSTRACT
Transmission of malaria parasites from humans to Anopheles mosquitoes can be inhibited by specific antibodies elicited during malaria infection, which target surface Plasmodium gametocyte/gamete proteins. Some of these proteins may have potential for vaccine development. Pvs48/45 is a P. vivax gametocyte surface antigen orthologous to Pfs48/45, which may play a role during parasite fertilization and thus has potential for transmission blocking (TB) activity. Here we describe the expression of a recombinant Pvs48/45 protein expressed in Escherichia coli as a â¼60kDa construct which we tested for antigenicity using human sera and for its immunogenicity and transmission blocking activity of specific anti-mouse and anti-monkey Pvs48/45 antibodies. The protein reacted with sera of individuals from malaria-endemic areas and in addition induced specific IgG antibody responses in BALB/c mice and Aotus l. griseimembra monkeys. Sera from both immunized animal species recognized native P. vivax protein in Western blot (WB) and immunofluorescence assays. Moreover, sera from immunized mice and monkeys produced significant inhibition of parasite transmission to An. Albimanus mosquitoes as shown by membrane feeding assays. Results indicate the presence of reactive epitopes in the Pvs48/45 recombinant product that induce antibodies with TB activity. Further testing of this protein is ongoing to determine its vaccine potential.
Subject(s)
Anopheles/immunology , Antigens, Protozoan/immunology , Malaria Vaccines/administration & dosage , Malaria, Vivax/prevention & control , Malaria, Vivax/transmission , Plasmodium vivax/genetics , Animals , Anopheles/parasitology , Antibodies, Protozoan/metabolism , Antigens, Protozoan/genetics , Aotidae/immunology , Aotidae/parasitology , Epitopes/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Haplorhini , Humans , Immunoglobulin G/metabolism , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Vivax/veterinary , Male , Mice , Mice, Inbred BALB C , Plasmodium vivax/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunologyABSTRACT
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/immunologyABSTRACT
BACKGROUND: Plasmodium vivax circumsporozoite (PvCS) protein is a major sporozoite surface antigen involved in parasite invasion of hepatocytes and is currently being considered as vaccine candidate. PvCS contains a dimorphic central repetitive fragment flanked by conserved regions that contain functional domains. METHODS: We have developed a chimeric 137-mer synthetic polypeptide (PvCS-NRC) that includes the conserved region I and region II-plus and the two natural repeat variants known as VK210 and VK247. The antigenicity of PvCS-NRC was tested using human sera from PNG and Colombia endemic areas and its immunogenicity was confirmed in mice with different genetic backgrounds, the polypeptide formulated either in Alum or GLA-SE adjuvants was assessed in inbred C3H, CB6F1 and outbred ICR mice, whereas a formulation in Montanide ISA51 was tested in C3H mice. RESULTS: Antigenicity studies indicated that the chimeric peptide is recognized by a high proportion (60-70%) of residents of malaria-endemic areas. Peptides formulated with either GLA-SE or Montanide ISA51 adjuvants induced stronger antibody responses as compared with the Alum formulation. Sera from immunized mice as well as antigen-specific affinity purified human IgG antibodies reacted with sporozoite preparations in immunofluorescence and Western blot assays, and displayed strong in vitro inhibition of sporozoite invasion (ISI) into hepatoma cells. CONCLUSIONS: The polypeptide was recognized at high prevalence when tested against naturally induced human antibodies and was able to induce significant immunogenicity in mice. Additionally, specific antibodies were able to recognize sporozoites and were able to block sporozoite invasion in vitro. Further evaluation of this chimeric protein construct in preclinical phase e.g. in Aotus monkeys in order to assess the humoral and cellular immune responses as well as protective efficacy against parasite challenge of the vaccine candidate must be conducted.
Subject(s)
Malaria Vaccines/immunology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Adult , Animals , Antibodies, Protozoan/blood , Blotting, Western , Cell Line , Colombia , Fluorescent Antibody Technique , Hepatocytes/parasitology , Humans , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mice , Plasmodium vivax/genetics , Protozoan Proteins/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunologyABSTRACT
A Plasmodium falciparum circumsporozoite protein (CSP)-based recombinant fusion vaccine is the first malaria vaccine to reach phase III clinical trials. Resistance to infection correlated with the production of antibodies to the immunodominant central repeat region of the CSP. In contrast to P. falciparum, vaccine development against the CSP of Plasmodium vivax malaria is far behind. Based on this gap in our knowledge, we generated a recombinant chimeric protein containing the immunodominant central repeat regions of the P. vivax CSP fused to Salmonella enterica serovar Typhimurium-derived flagellin (FliC) to activate the innate immune system. The recombinant proteins that were generated contained repeat regions derived from each of the 3 different allelic variants of the P. vivax CSP or a fusion of regions derived from each of the 3 allelic forms. Mice were subcutaneously immunized with the fusion proteins alone or in combination with the Toll-like receptor 3 (TLR-3) agonist poly(I·C), and the anti-CSP serum IgG response was measured. Immunization with a mixture of the 3 recombinant proteins, each containing immunodominant epitopes derived from a single allelic variant, rather than a single recombinant protein carrying a fusion of regions derived from each of 3 allelic forms elicited a stronger immune response. This response was independent of TLR-4 but required TLR-5/MyD88 activation. Antibody titers significantly increased when poly(I·C) was used as an adjuvant with a mixture of the 3 recombinant proteins. These recombinant fusion proteins are novel candidates for the development of an effective malaria vaccine against P. vivax.
Subject(s)
Adjuvants, Immunologic/pharmacology , Flagellin/pharmacology , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Salmonella typhimurium/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Animals , Antibodies, Protozoan/blood , Epitopes/genetics , Epitopes/immunology , Female , Flagellin/genetics , Injections, Subcutaneous , Malaria Vaccines/genetics , Mice , Plasmodium vivax/genetics , Poly I-C/administration & dosage , Poly I-C/pharmacology , Protozoan Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Salmonella typhimurium/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunologyABSTRACT
An important step when designing a vaccine is identifying the antigens that function as targets of naturally acquired antibodies. We investigated specific antibody responses against two Plasmodium vivax vaccine candidates, PvMSP-119 and PvMSP-3α359â798. Moreover, we assessed the relationship between these antibodies and morbidity parameters. PvMSP-119 was the most immunogenic antigen and the frequency of responders to this protein tended to increase in P. vivax patients with higher parasitemia. For both antigens, IgG antibody responses tended to be lower in patients who had experienced their first bout of malaria. Furthermore, anemic patients presented higher IgG antibody responses to PvMSP-3α359â798. Since the humoral response involves a number of antibodies acting simultaneously on different targets, we performed a Principal Component Analysis (PCA). Anemic patients had, on average, higher first principal component scores (IgG1/IgG2/IgG3/IgG4 anti-MSP3α), which were negatively correlated with hemoglobin levels. Since antibodies against PfMSP-3 have been strongly associated with clinical protection, we cannot exclude the possibility of a dual role of PvMSP-3 specific antibodies in both immunity and pathogenesis of vivax malaria. Our results confirm the high immunogenicity of the conserved C terminus of PvMSP-1 and points to the considerable immunogenicity of polymorphic PvMSP-3α359â798 during natural infection.
Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Vivax/immunology , Merozoite Surface Protein 1/immunology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Adolescent , Adult , Aged , Brazil , Child , Female , Humans , Immunoglobulin G/blood , Malaria Vaccines/administration & dosage , Male , Middle Aged , Young AdultABSTRACT
Latin America contributes 1-1.2 million clinical malaria cases to the global malaria burden of about 300 million per year. In 21 malaria endemic countries, the population at risk in this region represents less than 10% of the total population exposed worldwide. Factors such as rapid deforestation, inadequate agricultural practices, climate change, political instability, and both increasing parasite drug resistance and vector resistance to insecticides contribute to malaria transmission. Recently, several malaria endemic countries have experienced a significant reduction in numbers of malaria cases. This is most likely due to actions taken by National Malaria Control Programs (NMCP) with the support from international funding agencies. We describe here the research strategies and activities to be undertaken by the Centro Latino Americano de Investigación en Malaria (CLAIM), a new research center established for the non-Amazonian region of Latin America by the National Institute of Allergy and Infectious Diseases (NIAID). Throughout a network of countries in the region, initially including Colombia, Guatemala, Panama, and Peru, CLAIM will address major gaps in our understanding of changing malaria epidemiology, vector biology and control, and clinical malaria mainly due to Plasmodium vivax. In close partnership with NMCPs, CLAIM seeks to conduct research on how and why malaria is decreasing in many countries of the region as a basis for developing and implementing new strategies that will accelerate malaria elimination.
Subject(s)
Disease Eradication/methods , Disease Eradication/organization & administration , Epidemiologic Research Design , Malaria/prevention & control , Animals , Delivery of Health Care/organization & administration , Drug Resistance , Genetic Variation , Humans , Imidazoles/pharmacology , Insect Vectors/parasitology , Insect Vectors/physiology , International Cooperation , Latin America/epidemiology , Malaria/epidemiology , Malaria/immunology , Malaria/parasitology , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , National Health Programs/organization & administration , Niacin/analogs & derivatives , Niacin/pharmacology , Plasmodium/drug effects , Plasmodium/genetics , Plasmodium/immunology , Plasmodium/pathogenicity , Socioeconomic FactorsABSTRACT
Apical membrane antigen 1 (AMA-1) is an invasion-related Plasmodium antigen that is expressed during both intracellular and extracellular asexual stages of the parasite's life cycle, making it an ideal target for induction of humoral and cellular immune responses that can protect against malaria. We show here that when it is administered as a recombinant protein (P) in Montanide ISA720 adjuvant, followed by a recombinant human type 5 adenovirus (Ad), intense and long-lasting Plasmodium vivax AMA-1-specific antibody responses (including both IgG1 and IgG2a), as well as proliferative memory T cell responses, can be detected in immunized mice. Memory T cells displayed both central (CD44(hi) CD62L(hi)) and effector (CD44(hi) CD62L(lo)) phenotypes, with the central memory phenotype prevailing (56% of AMA-1-specific proliferating cells). Considering the main traits of the memory immune responses induced against AMA-1, this particular sequence of immunogens (P followed by Ad), but no others (Ad/Ad, Ad/P, or P/P), displayed an optimal synergistic effect. These results give further support to the need for preclinical studies of P. vivax vaccine candidate AMA-1 administered in prime/boost protocols that include recombinant proteins and adenoviral vectors.
Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Plasmodium vivax/immunology , Adenoviridae , Adjuvants, Immunologic , Animals , Antibodies, Protozoan/biosynthesis , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Hyaluronan Receptors/biosynthesis , Immunity, Cellular , Immunity, Humoral , Immunization , Immunization, Secondary , Immunologic Memory , L-Selectin/biosynthesis , Malaria Vaccines/administration & dosage , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Mannitol/immunology , Mice , Mice, Inbred BALB C , Oleic Acids/administration & dosage , Oleic Acids/immunology , Protozoan Proteins/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , T-Lymphocytes/immunology , Vaccines, Synthetic/immunologyABSTRACT
Mouse hepatitis virus A59 (MHV A59) induces autoantibodies (autoAb) to fumarylacetoacetate hydrolase (FAH), a soluble cytosolic enzyme present in the liver and kidneys, in various mouse strains. The aim of this work was to amplify and diversify the autoimmune response restricted to FAH through the use of the exogenous adjuvant called PADRE. Accordingly, C57BL/6 mice were chosen, because these animals respond to PADRE better than other mouse strains. Results presented herein indicate that, surprisingly, C57BL/6 mice developed signs of autoimmune hepatitis-like disease (AIH), including transient hypergammaglobulinemia, elevated transaminases, autoAb directed against different liver proteins and hepatic cellular infiltrates, indicating that a new model of experimental AIH could be generated by a viral inoculation. Furthermore, PADRE administration amplified the MHV effect, extending the duration of hypergammaglobulinemia and increasing the binding of autoAb as well as the degree of hepatic infiltrates. However, the adjuvant did not expand the time of the symptoms. Additionally, since plasmatic uric acid and high-mobility group box protein 1 (HGMB1) concentrations augmented in MHV- and/or PADRE-treated mice, it is suggested that both alarmins were probably involved in the spreading of the immune response induced by the viral infection and the adjuvant administration.
Subject(s)
Adjuvants, Immunologic/administration & dosage , Coronavirus Infections/immunology , Hepatitis, Viral, Animal/immunology , Malaria Vaccines/administration & dosage , Murine hepatitis virus/immunology , Animals , Autoantibodies/metabolism , Autoimmune Diseases/immunology , Disease Models, Animal , Disease Susceptibility , Hepatitis/immunology , Humans , Hydrolases/immunology , Hypergammaglobulinemia , Mice , Mice, Inbred C57BL , Murine hepatitis virus/pathogenicity , Transaminases/genetics , Transaminases/metabolismABSTRACT
In a recent study, we demonstrated the immunogenic properties of a new malaria vaccine polypeptide based on a 19 kDa C-terminal fragment of the merozoite surface protein-1 (MSP1(19)) from Plasmodium vivax and an innate immunity agonist, the Salmonella enterica serovar Typhimurium flagellin (FliC). Herein, we tested whether the same strategy, based on the MSP1(19) component of the deadly malaria parasite Plasmodium falciparum, could also generate a fusion polypeptide with enhanced immunogenicity. The His(6)FliC-MSP1(19) fusion protein was expressed from a recombinant Escherichia coli and showed preserved in vitro TLR5-binding activity. In contrast to animals injected with His(6)MSP1(19), mice subcutaneously immunised with the recombinant His(6)FliC-MSP1(19) developed strong MSP1(19)-specific systemic antibody responses with a prevailing IgG1 subclass. Incorporation of other adjuvants, such as CpG ODN 1826, complete and incomplete Freund's adjuvants or Quil-A, improved the IgG responses after the second, but not the third, immunising dose. It also resulted in a more balanced IgG subclass response, as evaluated by the IgG1/IgG2c ratio, and higher cell-mediated immune response, as determined by the detection of antigen-specific interferon-gamma secretion by immune spleen cells. MSP1(19)-specific antibodies recognised not only the recombinant protein, but also the native protein expressed on the surface of P. falciparum parasites. Finally, sera from rabbits immunised with the fusion protein alone inhibited the in vitro growth of three different P. falciparum strains. In summary, these results extend our previous observations and further demonstrate that fusion of the innate immunity agonist FliC to Plasmodium antigens is a promising alternative to improve their immunogenicity.
Subject(s)
Flagellin/immunology , Malaria Vaccines/immunology , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/immunology , Salmonella typhimurium/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/blood , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Escherichia coli/genetics , Female , Flagellin/genetics , Gene Expression , Immunoglobulin G/blood , Injections, Subcutaneous , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Merozoite Surface Protein 1/genetics , Mice , Mice, Inbred C57BL , Plasmodium falciparum/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Salmonella typhimurium/genetics , Spleen/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunologyABSTRACT
Despite the fact that we live in an era of advanced technology and innovation, infectious diseases, like malaria, continue to be one of the greatest health challenges worldwide. The main drawbacks of conventional malaria chemotherapy are the development of multiple drug resistance and the non-specific targeting to intracellular parasites, resulting in high dose requirements and subsequent intolerable toxicity. Nanosized carriers have been receiving special attention with the aim of minimizing the side effects of drug therapy, such as poor bioavailability and the selectivity of drugs. Several nanosized delivery systems have already proved their effectiveness in animal models for the treatment and prophylaxis of malaria. A number of strategies to deliver antimalarials using nanocarriers and the mechanisms that facilitate their targeting to Plasmodium spp.-infected cells are discussed in this review. Taking into account the peculiarities of malaria parasites, the focus is placed particularly on lipid-based (e.g., liposomes, solid lipid nanoparticles and nano and microemulsions) and polymer-based nanocarriers (nanocapsules and nanospheres). This review emphasizes the main requirements for developing new nanotechnology-based carriers as a promising choice in malaria treatment, especially in the case of severe cerebral malaria.
Subject(s)
Antimalarials/administration & dosage , Antimalarials/therapeutic use , Malaria Vaccines/administration & dosage , Malaria Vaccines/therapeutic use , Malaria/drug therapy , Nanotechnology/trends , Animals , Dendrimers , Developing Countries , Drug Carriers/chemistry , Drug Delivery Systems , Humans , Lipids/chemistry , Liposomes/chemistry , Malaria/parasitology , Malaria/prevention & control , Nanoparticles/chemistry , SuspensionsABSTRACT
The present study evaluated the immunogenicity of new malaria vaccine formulations based on the 19kDa C-terminal fragment of Plasmodium vivax Merozoite Surface Protein-1 (MSP1(19)) and the Salmonella enterica serovar Typhimurium flagellin (FliC), a Toll-like receptor 5 (TLR5) agonist. FliC was used as an adjuvant either admixed or genetically linked to the P. vivax MSP1(19) and administered to C57BL/6 mice via parenteral (s.c.) or mucosal (i.n.) routes. The recombinant fusion protein preserved MSP1(19) epitopes recognized by sera collected from P. vivax infected humans and TLR5 agonist activity. Mice parenterally immunized with recombinant P. vivax MSP1(19) in the presence of FliC, either admixed or genetically linked, elicited strong and long-lasting MSP1(19)-specific systemic antibody responses with a prevailing IgG1 subclass response. Incorporation of another TLR agonist, CpG ODN 1826, resulted in a more balanced response, as evaluated by the IgG1/IgG2c ratio, and higher cell-mediated immune response measured by interferon-gamma secretion. Finally, we show that MSP1(19)-specific antibodies recognized the native protein expressed on the surface of P. vivax parasites harvested from infected humans. The present report proposes a new class of malaria vaccine formulation based on the use of malarial antigens and the innate immunity agonist FliC. It contains intrinsic adjuvant properties and enhanced ability to induce specific humoral and cellular immune responses when administered alone or in combination with other adjuvants.
Subject(s)
Adjuvants, Immunologic , Flagellin/pharmacology , Malaria Vaccines/immunology , Merozoite Surface Protein 1/immunology , Plasmodium vivax/immunology , Salmonella typhimurium/metabolism , Toll-Like Receptor 5/agonists , Administration, Intranasal , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Chemistry, Pharmaceutical , Female , Flagellin/isolation & purification , Fluorescent Antibody Technique, Indirect , Immunization Schedule , Injections, Subcutaneous , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Merozoite Surface Protein 1/genetics , Mice , Mice, Inbred C57BL , Plasmodium vivax/genetics , Th1 Cells/immunology , Th2 Cells/immunology , Vaccines, Synthetic/immunologyABSTRACT
The recombinant apical membrane antigen 1 (AMA-1) and 19-kDa fragment of merozoite surface protein (MSP-1(19)) are the lead candidates for inclusion in a vaccine against blood stages of malaria due to encouraging protective studies in humans and animals. Despite the importance of an efficacious malaria vaccine, vaccine-related research has focused on identifying antigens that result in protective immunity rather than determining the nature of anti-malarial immune effector mechanisms. Moreover, emphasis has been placed on adaptive rather than innate immune responses. In this study, we investigated the effect of Plasmodium vivax vaccine candidates Pv-AMA-1 and Pv-MSP-1(19) on the immune response of malaria-naïve donors. Maturation of dendritic cells is altered by Pv-AMA-1 but not Pv-MSP-1(19), as observed by differential expression of cell surface markers. In addition, Pv-AMA-1 induced an increased production of MIP-1alpha/CCL3 and decreased production of TARC/CCL17 levels in both dendritic cells (DCs) and peripheral blood mononuclear cells (PBMCs). Finally, a significant pro-inflammatory response was elicited by Pv-AMA-1-stimulated PBMCs. These results suggest that the recombinant vaccine candidate Pv-AMA-1 may play a direct role on innate immune response and might be involved in parasite destruction.
Subject(s)
Antigens, Protozoan/immunology , Immunity, Innate , Malaria Vaccines/immunology , Malaria, Vivax/immunology , Membrane Proteins/immunology , Merozoite Surface Protein 1/immunology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Vaccines, Synthetic/immunology , Adult , Animals , Antigens, Protozoan/genetics , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria, Vivax/parasitology , Malaria, Vivax/prevention & control , Membrane Proteins/genetics , Merozoite Surface Protein 1/genetics , Protozoan Proteins/genetics , Vaccines, Synthetic/administration & dosageABSTRACT
Background: Malaria remains a leading global health problem that requires the improved use of existing interventions and the accelerated development of new control methods. We aimed to assess the safety, immunogenicity, and initial efficacy of the malaria vaccine RTS,S/AS02D in infants in Africa. Methods: We did a phase I/IIb double-blind randomised trial of 214 infants in Mozambique. Infants were randomly assigned to receive three doses either of RTS,S/AS02D or the hepatitis B vaccine Engerix-B at ages 10 weeks, 14 weeks, and 18 weeks of age, as well as routine immunisation vaccines given at 8, 12, and 16 weeks of age. The primary endpoint was safety of the RTS,S/AS02D during the first 6 months of the study, and analysis was by intention to treat. Secondary endpoints included immunogenicity and analysis of new Plasmodium falciparum infections during a 3-month follow up after the third dose. Time to new infections in the per-protocol cohort were compared between groups using Cox regression models. This study is registered with ClinicalTrials.gov, number NCT00197028. Findings: There were 17 children (15.9%; 95% CI 9.5-24.2) with serious adverse events in each group. In the follow-up which ended on March 6, 2007, there were 31 serious adverse events in the RTS,S/AS02D group and 30 serious adverse events in the Engerix-B group, none of which were reported as related to vaccination. There were four deaths during this same follow-up period; all of them after the active detection of infection period had finished at study month 6 (two in RTSS/AS02D group and two in the Engerix-B group). RTS,S/AS02D induced high titres of anti-circumsporozoite antibodies. 68 first or only P falciparum infections were documented: 22 in the RTS,S/AS02D group and 46 in the control group. The adjusted vaccine efficacy was 65.9% (95% CI 42.6-79.8%, p<0.0001). Interpretation: The RTS,S/AS02D malaria vaccine was safe, well tolerated, and immunogenic in young infants. These findings set the stage for expanded phase III efficacy studies to confirm vaccine efficacy against clinical malaria disease