151
|
Hodgson SH, Ewer KJ, Bliss CM, Edwards NJ, Rampling T, Anagnostou NA, de Barra E, Havelock T, Bowyer G, Poulton ID, de Cassan S, Longley R, Illingworth JJ, Douglas AD, Mange PB, Collins KA, Roberts R, Gerry S, Berrie E, Moyle S, Colloca S, Cortese R, Sinden RE, Gilbert SC, Bejon P, Lawrie AM, Nicosia A, Faust SN, Hill AVS. Evaluation of the efficacy of ChAd63-MVA vectored vaccines expressing circumsporozoite protein and ME-TRAP against controlled human malaria infection in malaria-naive individuals. J Infect Dis 2014; 211:1076-86. [PMID: 25336730 PMCID: PMC4354983 DOI: 10.1093/infdis/jiu579] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background. Circumsporozoite protein (CS) is the antigenic target for RTS,S, the most advanced malaria vaccine to date. Heterologous prime-boost with the viral vectors simian adenovirus 63 (ChAd63)-modified vaccinia virus Ankara (MVA) is the most potent inducer of T-cells in humans, demonstrating significant efficacy when expressing the preerythrocytic antigen insert multiple epitope–thrombospondin-related adhesion protein (ME-TRAP). We hypothesized that ChAd63-MVA containing CS may result in a significant clinical protective efficacy. Methods. We conducted an open-label, 2-site, partially randomized Plasmodium falciparum sporozoite controlled human malaria infection (CHMI) study to compare the clinical efficacy of ChAd63-MVA CS with ChAd63-MVA ME-TRAP. Results. One of 15 vaccinees (7%) receiving ChAd63-MVA CS and 2 of 15 (13%) receiving ChAd63-MVA ME-TRAP achieved sterile protection after CHMI. Three of 15 vaccinees (20%) receiving ChAd63-MVA CS and 5 of 15 (33%) receiving ChAd63-MVA ME-TRAP demonstrated a delay in time to treatment, compared with unvaccinated controls. In quantitative polymerase chain reaction analyses, ChAd63-MVA CS was estimated to reduce the liver parasite burden by 69%–79%, compared with 79%–84% for ChAd63-MVA ME-TRAP. Conclusions. ChAd63-MVA CS does reduce the liver parasite burden, but ChAd63-MVA ME-TRAP remains the most promising antigenic insert for a vectored liver-stage vaccine. Detailed analyses of parasite kinetics may allow detection of smaller but biologically important differences in vaccine efficacy that can influence future vaccine development. Clinical Trials Registration. NCT01623557.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eoghan de Barra
- Jenner Institute Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tom Havelock
- NIHR Wellcome Trust Clinical Research Facility, University of Southampton and University Hospital Southampton NHS Foundation Trust
| | | | | | | | | | | | | | | | | | | | | | - Eleanor Berrie
- Clinical Biomanufacturing Facility, University of Oxford
| | - Sarah Moyle
- Clinical Biomanufacturing Facility, University of Oxford
| | | | | | - Robert E Sinden
- Jenner Institute Division of Cell and Molecular Biology, Imperial College London, United Kingdom
| | | | - Philip Bejon
- Centre for Geographical Medical Research (Coast), Kenya Medical Research Institute-Wellcome Trust, Kilifi
| | | | - Alfredo Nicosia
- Okairos, Rome CEINGE Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Italy
| | - Saul N Faust
- NIHR Wellcome Trust Clinical Research Facility, University of Southampton and University Hospital Southampton NHS Foundation Trust
| | | |
Collapse
|
152
|
Gazzinelli RT, Kalantari P, Fitzgerald KA, Golenbock DT. Innate sensing of malaria parasites. Nat Rev Immunol 2014; 14:744-57. [PMID: 25324127 DOI: 10.1038/nri3742] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Innate immune receptors have a key role in immune surveillance by sensing microorganisms and initiating protective immune responses. However, the innate immune system is a classic 'double-edged sword' that can overreact to pathogens, which can have deleterious effects and lead to clinical manifestations. Recent studies have unveiled the complexity of innate immune receptors that function as sensors of Plasmodium spp. in the vertebrate host. This Review highlights the cellular and molecular mechanisms by which Plasmodium infection is sensed by different families of innate immune receptors. We also discuss how these events mediate both host resistance to infection and the pathogenesis of malaria.
Collapse
Affiliation(s)
- Ricardo T Gazzinelli
- 1] Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, 01605-02324 Worcester, Massachusetts, USA. [2] Laboratório de Imunopatologia, Centro de Pesquisa René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, Minas Gerais, Brazil. [3] Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Parisa Kalantari
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, 01605-02324 Worcester, Massachusetts, USA
| | - Katherine A Fitzgerald
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, 01605-02324 Worcester, Massachusetts, USA
| | - Douglas T Golenbock
- 1] Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, 01605-02324 Worcester, Massachusetts, USA. [2] Laboratório de Imunopatologia, Centro de Pesquisa René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
153
|
Ndeffo Mbah ML, Skrip L, Greenhalgh S, Hotez P, Galvani AP. Impact of Schistosoma mansoni on malaria transmission in Sub-Saharan Africa. PLoS Negl Trop Dis 2014; 8:e3234. [PMID: 25329403 PMCID: PMC4199517 DOI: 10.1371/journal.pntd.0003234] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 09/02/2014] [Indexed: 01/22/2023] Open
Abstract
Background Sub-Saharan Africa harbors the majority of the global burden of malaria and schistosomiasis infections. The co-endemicity of these two tropical diseases has prompted investigation into the mechanisms of coinfection, particularly the competing immunological responses associated with each disease. Epidemiological studies have shown that infection with Schistosoma mansoni is associated with a greater malaria incidence among school-age children. Methodology We developed a co-epidemic model of malaria and S. mansoni transmission dynamics which takes into account key epidemiological interaction between the two diseases in terms of elevated malaria incidence among individuals with S. mansoni high egg output. The model was parameterized for S. mansoni high-risk endemic communities, using epidemiological and clinical data of the interaction between S. mansoni and malaria among children in sub-Saharan Africa. We evaluated the potential impact of the S. mansoni–malaria interaction and mass treatment of schistosomiasis on malaria prevalence in co-endemic communities. Principal Findings Our results suggest that in the absence of mass drug administration of praziquantel, the interaction between S. mansoni and malaria may reduce the effectiveness of malaria treatment for curtailing malaria transmission, in S. mansoni high-risk endemic communities. However, when malaria treatment is used in combination with praziquantel, mass praziquantel administration may increase the effectiveness of malaria control intervention strategy for reducing malaria prevalence in malaria- S. mansoni co-endemic communities. Conclusions/Significance Schistosomiasis treatment and control programmes in regions where S. mansoni and malaria are highly prevalent may have indirect benefits on reducing malaria transmission as a result of disease interactions. In particular, mass praziquantel administration may not only have the direct benefit of reducing schistosomiasis infection, it may also reduce malaria transmission and disease burden. Malaria and Schistosoma mansoni are co-endemic in many regions of sub-Saharan Africa. Evidence from clinical and epidemiological studies support the hypothesis that concurrent infection with S. mansoni is associated with greater malaria incidence among school-age children. We use mathematical modeling to evaluate the epidemiological impact of S. mansoni infection on malaria transmission in sub-Saharan Africa. Using epidemiological data on the increased risk of malaria incidence in S. mansoni endemic communities from Senegal, we developed a co-epidemic model of malaria and S. mansoni transmission dynamics to address key epidemiological interactions between the two diseases. Parameterizing our model for S. mansoni high-risk endemic communities, we show that the interaction between S. mansoni and malaria may reduce the effectiveness of malaria treatment for curtailing malaria transmission. Moreover, we show that in addition to reducing schistosomiasis health burden, mass praziquantel administration will generate indirect benefit in terms of reducing malaria transmission and disease burden in S. mansoni–malaria co-endemic communities. Our findings indicate the possible benefit of scaling up schistosomiasis control efforts in sub-Saharan Africa, and especially in areas were S. mansoni and malaria are highly prevalent.
Collapse
Affiliation(s)
- Martial L. Ndeffo Mbah
- School of Public Health, Yale University, New Haven, Connecticut, United States of America
- * E-mail:
| | - Laura Skrip
- School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Scott Greenhalgh
- School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Peter Hotez
- National School of Tropical Medicine, and Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alison P. Galvani
- School of Public Health, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
154
|
Comer E, Beaudoin JA, Kato N, Fitzgerald ME, Heidebrecht RW, Lee MD, Masi D, Mercier M, Mulrooney C, Muncipinto G, Rowley A, Crespo-Llado K, Serrano AE, Lukens AK, Wiegand RC, Wirth DF, Palmer MA, Foley MA, Munoz B, Scherer CA, Duvall JR, Schreiber SL. Diversity-oriented synthesis-facilitated medicinal chemistry: toward the development of novel antimalarial agents. J Med Chem 2014; 57:8496-502. [PMID: 25211597 PMCID: PMC4207553 DOI: 10.1021/jm500994n] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
Here, we describe medicinal chemistry
that was accelerated by a
diversity-oriented synthesis (DOS) pathway, and in vivo studies of our previously reported macrocyclic antimalarial agent
that derived from the synthetic pathway. Structure–activity
relationships that focused on both appendage and skeletal features
yielded a nanomolar inhibitor of P. falciparum asexual
blood-stage growth with improved solubility and microsomal stability
and reduced hERG binding. The build/couple/pair (B/C/P) synthetic
strategy, used in the preparation of the original screening library,
facilitated medicinal chemistry optimization of the antimalarial lead.
Collapse
Affiliation(s)
- Eamon Comer
- Center for the Science of Therapeutics, Broad Institute , 7 Cambridge Center, Cambridge, Massachusetts 02142, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Ndure J, Flanagan KL. Targeting regulatory T cells to improve vaccine immunogenicity in early life. Front Microbiol 2014; 5:477. [PMID: 25309517 PMCID: PMC4161046 DOI: 10.3389/fmicb.2014.00477] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/25/2014] [Indexed: 12/26/2022] Open
Abstract
Human newborns and infants are bombarded with multiple pathogens on leaving the sterile intra-uterine environment, and yet have suboptimal innate immunity and limited immunological memory, thus leading to increased susceptibility to infections in early life. They are thus the target age group for a host of vaccines against common bacterial and viral pathogens. They are also the target group for many vaccines in development, including those against tuberculosis (TB), malaria, and HIV infection. However, neonatal and infant responses too many vaccines are suboptimal, and in the case of the polysaccharide vaccines, it has been necessary to develop the alternative conjugated formulations in order to induce immunity in early life. Immunoregulatory factors are an intrinsic component of natural immunity necessary to dampen or control immune responses, with the caveat that they may also decrease immunity to infections or lead to chronic infection. This review explores the key immunoregulatory factors at play in early life, with a particular emphasis on regulatory T cells (Tregs). It goes on to explore the role that Tregs play in limiting vaccine immunogenicity, and describes animal and human studies in which Tregs have been depleted in order to enhance vaccine responses. A deeper understanding of the role that Tregs play in limiting or controlling vaccine-induced immunity would provide strategies to improve vaccine immunogenicity in this critical age group. New adjuvants and drugs are being developed that can transiently suppress Treg function, and their use as part of human vaccination strategies against infections is becoming a real prospect for the future.
Collapse
Affiliation(s)
- Jorjoh Ndure
- Infant Immunology Group, Vaccinology Theme, Medical Research Council Laboratories Fajara, The Gambia
| | - Katie L Flanagan
- Vaccine and Infectious Diseases Laboratory, Department of Immunology, Monash University Melbourne, VIC, Australia
| |
Collapse
|
156
|
Doolan DL, Apte SH, Proietti C. Genome-based vaccine design: the promise for malaria and other infectious diseases. Int J Parasitol 2014; 44:901-13. [PMID: 25196370 DOI: 10.1016/j.ijpara.2014.07.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 01/08/2023]
Abstract
Vaccines are one of the most effective interventions to improve public health, however, the generation of highly effective vaccines for many diseases has remained difficult. Three chronic diseases that characterise these difficulties include malaria, tuberculosis and HIV, and they alone account for half of the global infectious disease burden. The whole organism vaccine approach pioneered by Jenner in 1796 and refined by Pasteur in 1857 with the "isolate, inactivate and inject" paradigm has proved highly successful for many viral and bacterial pathogens causing acute disease but has failed with respect to malaria, tuberculosis and HIV as well as many other diseases. A significant advance of the past decade has been the elucidation of the genomes, proteomes and transcriptomes of many pathogens. This information provides the foundation for new 21st Century approaches to identify target antigens for the development of vaccines, drugs and diagnostic tests. Innovative genome-based vaccine strategies have shown potential for a number of challenging pathogens, including malaria. We advocate that genome-based rational vaccine design will overcome the problem of poorly immunogenic, poorly protective vaccines that has plagued vaccine developers for many years.
Collapse
Affiliation(s)
- Denise L Doolan
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia.
| | - Simon H Apte
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Carla Proietti
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| |
Collapse
|
157
|
Sahi S, Rai S, Chaudhary M, Nain V. Modeling of human M1 aminopeptidases for in silico screening of potential Plasmodium falciparum alanine aminopeptidase (PfA-M1) specific inhibitors. Bioinformation 2014; 10:518-25. [PMID: 25258488 PMCID: PMC4166772 DOI: 10.6026/97320630010518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 06/27/2014] [Indexed: 11/23/2022] Open
Abstract
Plasmodium falciparum alanine M1-aminopeptidase (PfA-M1) is a validated target for anti-malarial drug development. Presence of significant similarity between PfA-M1 and human M1-aminopeptidases, particularly within regions of enzyme active site leads to problem of non-specificity and off-target binding for known aminopeptidase inhibitors. Molecular docking based in silico screening approach for off-target binding has high potential but requires 3D-structure of all human M1-aminopeptidaes. Therefore, in the present study 3D structural models of seven human M1-aminopeptidases were developed. The robustness of docking parameters and quality of predicted human M1-aminopeptidases structural models was evaluated by stereochemical analysis and docking of their respective known inhibitors. The docking scores were in agreement with the inhibitory concentrations elucidated in enzyme assays of respective inhibitor enzyme combinations (r2≈0.70). Further docking analysis of fifteen potential PfA-M1 inhibitors (virtual screening identified) showed that three compounds had less docking affinity for human M1-aminopeptidases as compared to PfA-M1. These three identified potential lead compounds can be validated with enzyme assays and used as a scaffold for designing of new compounds with increased specificity towards PfA-M1.
Collapse
Affiliation(s)
- Shakti Sahi
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201312, India
| | - Sneha Rai
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201312, India
| | - Meenakshi Chaudhary
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201312, India
| | - Vikrant Nain
- School of Biotechnology, Gautam Buddha University, Greater Noida, 201312, India
| |
Collapse
|
158
|
Vectored antibody gene delivery protects against Plasmodium falciparum sporozoite challenge in mice. Proc Natl Acad Sci U S A 2014; 111:12528-32. [PMID: 25114213 DOI: 10.1073/pnas.1407362111] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Malaria caused by Plasmodium falciparum kills nearly one million children each year and imposes crippling economic burdens on families and nations worldwide. No licensed vaccine exists, but infection can be prevented by antibodies against the circumsporozoite protein (CSP), the major surface protein of sporozoites, the form of the parasite injected by mosquitoes. We have used vectored immunoprophylaxis (VIP), an adeno-associated virus-based technology, to introduce preformed antibody genes encoding anti-P. falciparum CSP mAb into mice. VIP vector-transduced mice exhibited long-lived mAb expression at up to 1,200 µg/mL in serum, and up to 70% were protected from both i.v. and mosquito bite challenge with transgenic Plasmodium berghei rodent sporozoites that incorporate the P. falciparum target of the mAb in their CSP. Serum antibody levels and protection from mosquito bite challenge were dependent on the dose of the VIP vector. All individual mice expressing CSP-specific mAb 2A10 at 1 mg/mL or more were completely protected, suggesting that in this model system, exceeding that threshold results in consistent sterile protection. Our results demonstrate the potential of VIP as a path toward the elusive goal of immunization against malaria.
Collapse
|
159
|
Didierlaurent AM, Collignon C, Bourguignon P, Wouters S, Fierens K, Fochesato M, Dendouga N, Langlet C, Malissen B, Lambrecht BN, Garçon N, Van Mechelen M, Morel S. Enhancement of adaptive immunity by the human vaccine adjuvant AS01 depends on activated dendritic cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:1920-30. [PMID: 25024381 DOI: 10.4049/jimmunol.1400948] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Adjuvant System AS01 is a liposome-based vaccine adjuvant containing 3-O-desacyl-4'-monophosphoryl lipid A and the saponin QS-21. AS01 has been selected for the clinical development of several candidate vaccines including the RTS,S malaria vaccine and the subunit glycoprotein E varicella zoster vaccine (both currently in phase III). Given the known immunostimulatory properties of MPL and QS-21, the objective of this study was to describe the early immune response parameters after immunization with an AS01-adjuvanted vaccine and to identify relationships with the vaccine-specific adaptive immune response. Cytokine production and innate immune cell recruitment occurred rapidly and transiently at the muscle injection site and draining lymph node postinjection, consistent with the rapid drainage of the vaccine components to the draining lymph node. The induction of Ag-specific Ab and T cell responses was dependent on the Ag being injected at the same time or within 24 h after AS01, suggesting that the early events occurring postinjection were required for these elevated adaptive responses. In the draining lymph node, after 24 h, the numbers of activated and Ag-loaded monocytes and MHCII(high) dendritic cells were higher after the injection of the AS01-adjuvanted vaccine than after Ag alone. However, only MHCII(high) dendritic cells appeared efficient at and necessary for direct Ag presentation to T cells. These data suggest that the ability of AS01 to improve adaptive immune responses, as has been demonstrated in clinical trials, is linked to a transient stimulation of the innate immune system leading to the generation of high number of efficient Ag-presenting dendritic cells.
Collapse
Affiliation(s)
| | | | | | | | - Kaat Fierens
- Vlaams Instituut voor Biotechnologie Inflammation Research Center, Ghent University, 9052 Ghent, Belgium; and
| | | | | | | | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, INSERM U1104, Centre National de la Recherche Scientifique Unité Mixte de Recherche 7280, 13288 Marseille cedex 9, France
| | - Bart N Lambrecht
- Vlaams Instituut voor Biotechnologie Inflammation Research Center, Ghent University, 9052 Ghent, Belgium; and
| | | | | | - Sandra Morel
- GlaxoSmithKline Vaccines, 1330 Rixensart, Belgium
| |
Collapse
|
160
|
White MT, Bejon P, Olotu A, Griffin JT, Bojang K, Lusingu J, Salim N, Abdulla S, Otsyula N, Agnandji ST, Lell B, Asante KP, Owusu-Agyei S, Mahama E, Agbenyega T, Ansong D, Sacarlal J, Aponte JJ, Ghani AC. A combined analysis of immunogenicity, antibody kinetics and vaccine efficacy from phase 2 trials of the RTS,S malaria vaccine. BMC Med 2014; 12:117. [PMID: 25012228 PMCID: PMC4227280 DOI: 10.1186/s12916-014-0117-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/19/2014] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The RTS,S malaria vaccine is currently undergoing phase 3 trials. High vaccine-induced antibody titres to the circumsporozoite protein (CSP) antigen have been associated with protection from infection and episodes of clinical malaria. METHODS Using data from 5,144 participants in nine phase 2 trials, we explore predictors of vaccine immunogenicity (anti-CSP antibody titres), decay in antibody titres, and the association between antibody titres and clinical outcomes. We use empirically-observed relationships between these factors to predict vaccine efficacy in a range of scenarios. RESULTS Vaccine-induced anti-CSP antibody titres were significantly associated with age (P = 0.04), adjuvant (P <0.001), pre-vaccination anti-hepatitis B surface antigen titres (P = 0.005) and pre-vaccination anti-CSP titres (P <0.001). Co-administration with other vaccines reduced anti-CSP antibody titres although not significantly (P = 0.095). Antibody titres showed a bi-phasic decay over time with an initial rapid decay in the first three months and a second slower decay over the next three to four years. Antibody titres were significantly associated with protection, with a titre of 51 (95% Credible Interval (CrI): 29 to 85) ELISA units/ml (EU/mL) predicted to prevent 50% of infections in children. Vaccine efficacy was predicted to decline to zero over four years in a setting with entomological inoculation rate (EIR) = 20 infectious bites per year (ibpy). Over a five-year follow-up period at an EIR = 20 ibpy, we predict RTS,S will avert 1,782 cases per 1,000 vaccinated children, 1,452 cases per 1,000 vaccinated infants, and 887 cases per 1,000 infants when co-administered with expanded programme on immunisation (EPI) vaccines. Our main study limitations include an absence of vaccine-induced cellular immune responses and short duration of follow-up in some individuals. CONCLUSIONS Vaccine-induced anti-CSP antibody titres and transmission intensity can explain variations in observed vaccine efficacy.
Collapse
|
161
|
Miura K, Jongert E, Deng B, Zhou L, Lusingu JP, Drakeley CJ, Fay MP, Long CA, Vekemans J. Effect of ingested human antibodies induced by RTS, S/AS01 malaria vaccination in children on Plasmodium falciparum oocyst formation and sporogony in mosquitoes. Malar J 2014; 13:263. [PMID: 25007730 PMCID: PMC4100490 DOI: 10.1186/1475-2875-13-263] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 07/03/2014] [Indexed: 12/18/2022] Open
Abstract
Background The circumsporozoite protein (CS protein) on the malaria parasites in mosquitoes plays an important role in sporogony in mosquitoes. The RTS,S/AS01 malaria vaccine candidate, which has shown significant efficacy against clinical malaria in a large Phase 3 trial, targets the Plasmodium falciparum CS protein, but the ability of serum from vaccinated individuals to inhibit sporogony in mosquitoes has not been evaluated. Methods Previously a double-blind, randomized trial of RTS,S/AS01 vaccine, as compared with rabies vaccine, in five- to 17-month old children in Tanzania was conducted. In this study, polyclonal human antibodies were purified from the pools of sera taken one month after the third vaccination. IgGs were purified from four pools of sera from 25 RTS,S/AS01 vaccinated children each, and two pools of sera from 25 children vaccinated with rabies vaccine each. The ability of antibodies to inhibit P. falciparum oocyst formation and/or sporogony in the mosquito host was evaluated by a standard membrane-feeding assay. The test antibodies were fed on day 0 (at the same time as the gametocyte feed), or on days 3 or 6 (serial-feed experiments). The oocyst and sporozoite counts were performed on days 8 and 16, respectively. In addition, two human anti-CS monoclonal antibodies (mAb) and a control mAb were also evaluated. Results Polyclonal anti-CS IgG preparations from RTS,S-vaccinated children tested at concentrations of 149-210 ELISA units (EU)/ml did not show significant inhibition in oocyst and sporozoite formation when the antibodies were fed with gametocytes at the same time, or later (serial-feed experiments). Similarly, anti-CS mAbs tested at 6,421 or 7,122 EU/ml did not show reduction in oocyst and sporozoite formation. Conclusions This study does not support the concept that anti-CS antibodies induced by the RTS,S/AS01 vaccines in humans noticeably reduce malaria transmission by blocking P. falciparum sporozoite development or salivary gland invasion in mosquitoes when taken up during feeding.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Efficacy and safety of the RTS,S/AS01 malaria vaccine during 18 months after vaccination: a phase 3 randomized, controlled trial in children and young infants at 11 African sites. PLoS Med 2014; 11:e1001685. [PMID: 25072396 PMCID: PMC4114488 DOI: 10.1371/journal.pmed.1001685] [Citation(s) in RCA: 312] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 06/18/2014] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND A malaria vaccine could be an important addition to current control strategies. We report the safety and vaccine efficacy (VE) of the RTS,S/AS01 vaccine during 18 mo following vaccination at 11 African sites with varying malaria transmission. METHODS AND FINDINGS 6,537 infants aged 6-12 wk and 8,923 children aged 5-17 mo were randomized to receive three doses of RTS,S/AS01 or comparator vaccine. VE against clinical malaria in children during the 18 mo after vaccine dose 3 (per protocol) was 46% (95% CI 42% to 50%) (range 40% to 77%; VE, p<0.01 across all sites). VE during the 20 mo after vaccine dose 1 (intention to treat [ITT]) was 45% (95% CI 41% to 49%). VE against severe malaria, malaria hospitalization, and all-cause hospitalization was 34% (95% CI 15% to 48%), 41% (95% CI 30% to 50%), and 19% (95% CI 11% to 27%), respectively (ITT). VE against clinical malaria in infants was 27% (95% CI 20% to 32%, per protocol; 27% [95% CI 21% to 33%], ITT), with no significant protection against severe malaria, malaria hospitalization, or all-cause hospitalization. Post-vaccination anti-circumsporozoite antibody geometric mean titer varied from 348 to 787 EU/ml across sites in children and from 117 to 335 EU/ml in infants (per protocol). VE waned over time in both age categories (Schoenfeld residuals p<0.001). The number of clinical and severe malaria cases averted per 1,000 children vaccinated ranged across sites from 37 to 2,365 and from -1 to 49, respectively; corresponding ranges among infants were -10 to 1,402 and -13 to 37, respectively (ITT). Meningitis was reported as a serious adverse event in 16/5,949 and 1/2,974 children and in 9/4,358 and 3/2,179 infants in the RTS,S/AS01 and control groups, respectively. CONCLUSIONS RTS,S/AS01 prevented many cases of clinical and severe malaria over the 18 mo after vaccine dose 3, with the highest impact in areas with the greatest malaria incidence. VE was higher in children than in infants, but even at modest levels of VE, the number of malaria cases averted was substantial. RTS,S/AS01 could be an important addition to current malaria control in Africa. TRIAL REGISTRATION www.ClinicalTrials.gov NCT00866619 Please see later in the article for the Editors' Summary.
Collapse
|
163
|
Lorenz V, Karanis G, Karanis P. Malaria vaccine development and how external forces shape it: an overview. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2014; 11:6791-807. [PMID: 24983392 PMCID: PMC4113845 DOI: 10.3390/ijerph110706791] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 06/03/2014] [Accepted: 06/04/2014] [Indexed: 11/16/2022]
Abstract
The aim of this paper is to analyse the current status and scientific value of malaria vaccine approaches and to provide a realistic prognosis for future developments. We systematically review previous approaches to malaria vaccination, address how vaccine efforts have developed, how this issue may be fixed, and how external forces shape vaccine development. Our analysis provides significant information on the various aspects and on the external factors that shape malaria vaccine development and reveal the importance of vaccine development in our society.
Collapse
Affiliation(s)
- Veronique Lorenz
- Center of Anatomy, Medical School, University of Cologne, Cologne 50937, Germany.
| | - Gabriele Karanis
- National Research Center for Protozoan Diseases, Obihiro University for Agriculture and Veterinary Medicine, Hokkaido 080-8555, Japan.
| | - Panagiotis Karanis
- Center of Anatomy, Medical School, University of Cologne, Cologne 50937, Germany.
| |
Collapse
|
164
|
Nahrendorf W, Scholzen A, Bijker EM, Teirlinck AC, Bastiaens GJH, Schats R, Hermsen CC, Visser LG, Langhorne J, Sauerwein RW. Memory B-cell and antibody responses induced by Plasmodium falciparum sporozoite immunization. J Infect Dis 2014; 210:1981-90. [PMID: 24970846 PMCID: PMC4241945 DOI: 10.1093/infdis/jiu354] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Immunization of healthy volunteers during receipt of chemoprophylaxis with Plasmodium falciparum sporozoites (CPS-immunization) induces sterile protection from malaria. Antibody responses have long been known to contribute to naturally acquired immunity against malaria, but their association with sterile protection after whole sporozoite immunization is not well established. We therefore studied the induction and kinetics of malaria parasite antigen-specific antibodies and memory B-cells (MBCs) during CPS-immunization and their correlation with protection from challenge infection. Methods We assessed humoral reactivity to 9 antigens representing different stages of the life cycle of P. falciparum by performing standardized MBC enzyme-linked immunospot and enzyme-linked immunosorbent assays on peripheral blood mononuclear cells and plasma samples from 38 Dutch volunteers enrolled in 2 randomized controlled clinical trials. Results MBCs and antibodies recognizing pre-erythrocytic and cross-stage antigens were gradually acquired during CPS-immunization. The magnitude of these humoral responses did not correlate with protection but directly reflected parasite exposure in CPS-immunization and challenge. Conclusions Humoral responses to the malarial antigens circumsporozoite protein, liver-stage antigen-1, apical membrane antigen-1, and merozoite surface protein-1 do not to predict protection from challenge infection but can be used as sensitive marker of recent parasite exposure. Clinical Trials Registration NCT01236612 and NCT01218893.
Collapse
Affiliation(s)
- Wiebke Nahrendorf
- Department of Medical Microbiology, Radboud university medical center, Nijmegen Division of Parasitology, MRC National Institute for Medical Research, London, United Kingdom
| | - Anja Scholzen
- Department of Medical Microbiology, Radboud university medical center, Nijmegen
| | - Else M Bijker
- Department of Medical Microbiology, Radboud university medical center, Nijmegen
| | - Anne C Teirlinck
- Department of Medical Microbiology, Radboud university medical center, Nijmegen
| | - Guido J H Bastiaens
- Department of Medical Microbiology, Radboud university medical center, Nijmegen
| | - Remko Schats
- Department of Infectious Diseases, Leiden University Medical Center, The Netherlands
| | - Cornelus C Hermsen
- Department of Medical Microbiology, Radboud university medical center, Nijmegen
| | - Leo G Visser
- Department of Infectious Diseases, Leiden University Medical Center, The Netherlands
| | - Jean Langhorne
- Division of Parasitology, MRC National Institute for Medical Research, London, United Kingdom
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud university medical center, Nijmegen
| |
Collapse
|
165
|
Immunization with a functional protein complex required for erythrocyte invasion protects against lethal malaria. Proc Natl Acad Sci U S A 2014; 111:10311-6. [PMID: 24958881 DOI: 10.1073/pnas.1409928111] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
An essential step in the invasion of red blood cells (RBCs) by Plasmodium falciparum (Pf) merozoites is the binding of rhoptry neck protein 2 (RON2) to the hydrophobic groove of apical membrane antigen 1 (AMA1), triggering junction formation between the apical end of the merozoite and the RBC surface to initiate invasion. Vaccination with AMA1 provided protection against homologous parasites in one of two phase 2 clinical trials; however, despite its ability to induce high-titer invasion-blocking antibodies in a controlled human challenge trial, the vaccine conferred little protection even against the homologous parasite. Here we provide evidence that immunization with an AMA1-RON2 peptide complex, but not with AMA1 alone, provided complete protection against a lethal Plasmodium yoelii challenge in mice. Significantly, IgG from mice immunized with the complex transferred protection. Furthermore, IgG from PfAMA1-RON2-immunized animals showed enhanced invasion inhibition compared with IgG elicited by AMA1 alone. Interestingly, this qualitative increase in inhibitory activity appears to be related, at least in part, to a switch in the proportion of IgG specific for certain loop regions in AMA1 surrounding the binding site of RON2. Antibodies induced by the complex were not sufficient to block the FVO strain heterologous parasite, however, reinforcing the need to include multiallele AMA1 to cover polymorphisms. Our results suggest that AMA1 subunit vaccines may be highly effective when presented to the immune system as an invasion complex with RON2.
Collapse
|
166
|
Deng X, Kokkonda S, El Mazouni F, White J, Burrows JN, Kaminsky W, Charman SA, Matthews D, Rathod PK, Phillips MA. Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors. J Med Chem 2014; 57:5381-94. [PMID: 24801997 PMCID: PMC4079327 DOI: 10.1021/jm500481t] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Malaria is one of the most serious global infectious diseases. The pyrimidine biosynthetic enzyme Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) is an important target for antimalarial chemotherapy. We describe a detailed analysis of protein-ligand interactions between DHODH and a triazolopyrimidine-based inhibitor series to explore the effects of fluorine on affinity and species selectivity. We show that increasing fluorination dramatically increases binding to mammalian DHODHs, leading to a loss of species selectivity. Triazolopyrimidines bind Plasmodium and mammalian DHODHs in overlapping but distinct binding sites. Key hydrogen-bond and stacking interactions underlying strong binding to PfDHODH are absent in the mammalian enzymes. Increasing fluorine substitution leads to an increase in the entropic contribution to binding, suggesting that strong binding to mammalian DHODH is a consequence of an enhanced hydrophobic effect upon binding to an apolar pocket. We conclude that hydrophobic interactions between fluorine and hydrocarbons provide significant binding energy to protein-ligand interactions. Our studies define the requirements for species-selective binding to PfDHODH and show that the triazolopyrimidine scaffold can alternatively be tuned to inhibit human DHODH, an important target for autoimmune diseases.
Collapse
Affiliation(s)
- Xiaoyi Deng
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas , 6001 Forest Park Boulevard, Dallas, Texas 75390-9041, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Stoler J, Al Dashti R, Anto F, Fobil JN, Awandare GA. Deconstructing "malaria": West Africa as the next front for dengue fever surveillance and control. Acta Trop 2014; 134:58-65. [PMID: 24613157 DOI: 10.1016/j.actatropica.2014.02.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 02/19/2014] [Accepted: 02/23/2014] [Indexed: 11/15/2022]
Abstract
Presumptive treatment of febrile illness patients for malaria remains the norm in endemic areas of West Africa, and "malaria" remains the top source of health facility outpatient visits in many West African nations. Many other febrile illnesses, including bacterial, viral, and fungal infections, share a similar symptomatology as malaria and are routinely misdiagnosed as such; yet growing evidence suggests that much of the burden of febrile illness is often not attributable to malaria. Dengue fever is one of several viral diseases with symptoms similar to malaria, and the combination of rapid globalization, the long-standing presence of Aedes mosquitoes, case reports from travelers, and recent seroprevalence surveys all implicate West Africa as an emerging front for dengue surveillance and control. This paper integrates recent vector ecology, public health, and clinical medicine literature about dengue in West Africa across community, regional, and global geographic scales. We present a holistic argument for greater attention to dengue fever surveillance in West Africa and renew the call for improving differential diagnosis of febrile illness patients in the region.
Collapse
Affiliation(s)
- Justin Stoler
- Department of Geography and Regional Studies, University of Miami, 1300 Campo Sano Avenue, Coral Gables, FL, USA; Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Rawan Al Dashti
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Francis Anto
- Department of Epidemiology and Disease Control, University of Ghana, Legon, Ghana.
| | - Julius N Fobil
- Department of Biological, Environmental & Occupational Health Sciences, University of Ghana, Legon, Ghana.
| | - Gordon A Awandare
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Ghana.
| |
Collapse
|
168
|
de Souza JB. Protective immunity against malaria after vaccination. Parasite Immunol 2014; 36:131-9. [PMID: 24188045 DOI: 10.1111/pim.12086] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 10/29/2013] [Indexed: 11/28/2022]
Abstract
A good understanding of the immunological correlates of protective immunity is an important requirement for the development of effective vaccines against malaria. However, this concern has received little attention even in the face of two decades of intensive vaccine research. Here, we review the immune response to blood-stage malaria, with a particular focus on the type of vaccine most likely to induce the kind of response required to give strong protection against infection.
Collapse
Affiliation(s)
- J B de Souza
- Faculty of Infectious and Tropical Diseases, Department of Immunity and Infection, London School of Hygiene & Tropical Medicine, London, UK; Division of Infection & Immunity, University College London Medical School, London, UK
| |
Collapse
|
169
|
Wykes MN, Horne-Debets JM, Leow CY, Karunarathne DS. Malaria drives T cells to exhaustion. Front Microbiol 2014; 5:249. [PMID: 24904561 PMCID: PMC4034037 DOI: 10.3389/fmicb.2014.00249] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/07/2014] [Indexed: 01/08/2023] Open
Abstract
Malaria is a significant global burden but after >30 years of effort there is no vaccine on the market. While the complex life cycle of the parasite presents several challenges, many years of research have also identified several mechanisms of immune evasion by Plasmodium spp. Recent research on malaria, has investigated the programmed cell death-1 (PD-1) pathway which mediates exhaustion of T cells, characterized by poor effector functions and recall responses and in some cases loss of the cells by apoptosis. Such studies have shown exhaustion of CD4(+) T cells and an unappreciated role for CD8(+) T cells in promoting sterile immunity against blood stage malaria. This is because PD-1 mediates up to a 95% reduction in numbers and functional capacity of parasite-specific CD8(+) T cells, thus masking their role in protection. The role of T cell exhaustion during malaria provides an explanation for the absence of sterile immunity following the clearance of acute disease which will be relevant to future malaria-vaccine design and suggests the need for novel therapeutic solutions. This review will thus examine the role of PD-1-mediated T cell exhaustion in preventing lasting immunity against malaria.
Collapse
Affiliation(s)
- Michelle N Wykes
- Molecular Immunology Laboratory, QIMR Berghofer Medical Research Institute Brisbane, QLD, Australia
| | - Joshua M Horne-Debets
- Molecular Immunology Laboratory, QIMR Berghofer Medical Research Institute Brisbane, QLD, Australia ; The School of Medicine, University of Queensland Brisbane, QLD, Australia
| | - Chiuan-Yee Leow
- Molecular Immunology Laboratory, QIMR Berghofer Medical Research Institute Brisbane, QLD, Australia
| | | |
Collapse
|
170
|
Ojakaa DI, Jarvis JD, Matilu MI, Thiam S. Acceptance of a malaria vaccine by caregivers of sick children in Kenya. Malar J 2014; 13:172. [PMID: 24886650 PMCID: PMC4022976 DOI: 10.1186/1475-2875-13-172] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 05/01/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Several malaria vaccines are currently in clinical trials and are expected to provide an improved strategy for malaria control. Prior to introduction of a new vaccine, policymakers must consider the socio cultural environment of the region to ensure widespread community approval. This study investigated the acceptance of a malaria vaccine by child caregivers and analysed factors that influence these. METHODS Interviews from a standard questionnaire were conducted with 2,003 caregivers at 695 randomly selected health facilities across Kenya during the Kenya Service Provision Assessment Survey 2010. Multinomial regression of quantitative data was conducted using STATA to analyse determinants of caregivers accepting malaria vaccination of their child. RESULTS Mothers represented 90% of caregivers interviewed who brought their child to the health facility, and 77% of caregivers were 20-34 years old. Overall, 88% of respondents indicated that they would accept a malaria vaccine, both for a child in their community and their own child. Approval for a vaccine was highest in malaria-endemic Nyanza Province at 98.9%, and lowest in the seasonal transmission area of North Eastern Province at 23%. Although 94% of respondents who had attended at least some school reported they would accept the vaccine for a child, only 56% of those who had never attended school would do so. The likelihood of accepting one's own child to be immunized was correlated with province, satisfaction with health care services in the facility attended, age of the caregiver, and level of education. CONCLUSIONS Results from this study indicate a need for targeted messages and education on a malaria vaccine, particularly for residents of regions where acceptance is low, older caregivers, and those with low literacy and school-attendance levels. This study provides critical evidence to inform policy for a new malaria vaccine that will support its timely and comprehensive uptake in Kenya.
Collapse
|
171
|
Differing Patterns of Selection and Geospatial Genetic Diversity within Two Leading Plasmodium vivax Candidate Vaccine Antigens. PLoS Negl Trop Dis 2014; 8:e2796. [PMID: 24743266 PMCID: PMC3990511 DOI: 10.1371/journal.pntd.0002796] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 03/05/2014] [Indexed: 02/04/2023] Open
|
172
|
White MT, Griffin JT, Akpogheneta O, Conway DJ, Koram KA, Riley EM, Ghani AC. Dynamics of the Antibody Response to Plasmodium falciparum Infection in African Children. J Infect Dis 2014; 210:1115-22. [DOI: 10.1093/infdis/jiu219] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
173
|
Wang Y, Bhattacharya D. Adjuvant-specific regulation of long-term antibody responses by ZBTB20. ACTA ACUST UNITED AC 2014; 211:841-56. [PMID: 24711582 PMCID: PMC4010912 DOI: 10.1084/jem.20131821] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
ZBTB20 promotes long-lived plasma cell survival after primary activation under alum but not TLR2- and TLR4-activating adjuvant conditions. The duration of antibody production by long-lived plasma cells varies with the type of immunization, but the basis for these differences is unknown. We demonstrate that plasma cells formed in response to the same immunogen engage distinct survival programs depending on the adjuvant. After alum-adjuvanted immunization, antigen-specific bone marrow plasma cells deficient in the transcription factor ZBTB20 failed to accumulate over time, leading to a progressive loss of antibody production relative to wild-type controls. Fetal liver reconstitution experiments demonstrated that the requirement for ZBTB20 was B cell intrinsic. No defects were observed in germinal center numbers, affinity maturation, or plasma cell formation or proliferation in ZBTB20-deficient chimeras. However, ZBTB20-deficient plasma cells expressed reduced levels of MCL1 relative to wild-type controls, and transgenic expression of BCL2 increased serum antibody titers. These data indicate a role for ZBTB20 in promoting survival in plasma cells. Strikingly, adjuvants that activate TLR2 and TLR4 restored long-term antibody production in ZBTB20-deficient chimeras through the induction of compensatory survival programs in plasma cells. Thus, distinct lifespans are imprinted in plasma cells as they are formed, depending on the primary activation conditions. The durability of vaccines may accordingly be improved through the selection of appropriate adjuvants.
Collapse
Affiliation(s)
- Yinan Wang
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110
| | | |
Collapse
|
174
|
Behet MC, Foquet L, van Gemert GJ, Bijker EM, Meuleman P, Leroux-Roels G, Hermsen CC, Scholzen A, Sauerwein RW. Sporozoite immunization of human volunteers under chemoprophylaxis induces functional antibodies against pre-erythrocytic stages of Plasmodium falciparum. Malar J 2014; 13:136. [PMID: 24708526 PMCID: PMC4113136 DOI: 10.1186/1475-2875-13-136] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/26/2014] [Indexed: 11/24/2022] Open
Abstract
Background Long-lasting and sterile protective immunity against Plasmodium falciparum can be achieved by immunization of malaria-naive human volunteers under chloroquine prophylaxis with sporozoites delivered by mosquito bites (CPS-immunization). Protection is mediated by sporozoite/liver-stage immunity. In this study, the capacity of CPS-induced antibodies to interfere with sporozoite functionality and development was explored. Methods IgG was purified from plasma samples obtained before and after CPS-immunization from two separate clinical trials. The functionality of these antibodies was assessed in vitro in gliding and human hepatocyte traversal assays, and in vivo in a human liver-chimeric mouse model. Results Whereas pre-treatment of sporozoites with 2 mg/ml IgG in the majority of the volunteers did not have an effect on in vitro sporozoite gliding motility, CPS-induced IgG showed a distinct inhibitory effect in the sporozoite in vitro traversal assay. Pre-treatment of P. falciparum sporozoites with post-immunization IgG significantly inhibited sporozoite traversal through hepatocytes in 9/9 samples when using 10 and 1 mg/ml IgG, and was dose-dependent, resulting in an average 16% and 37% reduction with 1 mg/ml IgG (p = 0.003) and 10 mg/ml IgG (p = 0.002), respectively. In vivo, CPS-induced IgG reduced liver-stage infection and/or development after a mosquito infection in the human liver-chimeric mouse model by 91.05% when comparing 11 mice receiving post-immunization IgG to 11 mice receiving pre-immunization IgG (p = 0.0008). Conclusions It is demonstrated for the first time that CPS-immunization induces functional antibodies against P. falciparum sporozoites, which are able to reduce parasite-host cell interaction by inhibiting parasite traversal and liver-stage infection. These data highlight the functional contribution of antibody responses to pre-erythrocytic immunity after whole-parasite immunization against P. falciparum malaria.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Robert W Sauerwein
- Radboud University Medical Center, Department of Medical Microbiology, Geert Grooteplein 28, Microbiology 268, Nijmegen, HB 6500, The Netherlands.
| |
Collapse
|
175
|
Duration of vaccine efficacy against malaria: 5th year of follow-up in children vaccinated with RTS,S/AS02 in Mozambique. Vaccine 2014; 32:2209-16. [PMID: 24631081 DOI: 10.1016/j.vaccine.2014.02.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 12/18/2013] [Accepted: 02/12/2014] [Indexed: 02/07/2023]
Abstract
A primary concern for the RTS,S malaria vaccine candidate is duration of protection. The ongoing Phase III trial reported evidence of waning efficacy within the first year following vaccination. Multiple Phase IIb trials demonstrated early waning of efficacy. The longest duration of protection for RTS,S recorded to date was in a trial of a cohort of 1605 Mozambican children age 1-4 yr at the time of immunization (C1), which showed an overall efficacy against clinical malaria of 30.5% over 43 subsequent months of surveillance. A significant reduction in parasite prevalence in RTS,S vaccinees indicated that the vaccine continued to protect at the end of this period. Although follow-up for recording incident cases of clinical malaria was stopped at 45 months, we were interested in evidence of further durability of protection, and revisited the cohort at 63 months, recording the secondary trial endpoint, prevalence of asexual Plasmodium falciparum parasitemia, in the RTS,S and comparator vaccine groups as a proxy for efficacy. As a comparator, we also visited the contemporaneous cohort of 417 children (C2), which showed waning efficacy after 6 months of follow-up. We also assessed anti-circumsporozoite antibody titers. These results were compared with those of other Phase IIb trials. Prevalence of parasitemia was not significantly lower in the RTS,S/AS02 group compared to comparator groups in C1 (57 [119%] Vs 62 [128%]; p=0.696) or C2 (30 [226%] Vs 35 [276%]; p=0.391), despite elevated antibody titers, suggesting that protection did not extend to 5 years after vaccination. This is in contrast to the earlier assessment of parasitemia in C1, where a 34% lower prevalence of parasitemia was observed in the RTS,S/AS02 group at month 45. Comparison with other Phase II trials highlights a complex relationship between efficacy, age and transmission intensity. RTS,S/AS02 provided partial protection from clinical malaria for at least 3.5 years in C1. Duration of protection may depend on environmental circumstances, such as changing malaria transmission, and special attention should be given in the Phase III trial to identifying factors that modify longevity of protection.
Collapse
|
176
|
The whole parasite, pre-erythrocytic stage approach to malaria vaccine development: a review. Curr Opin Infect Dis 2014; 26:420-8. [PMID: 23982233 DOI: 10.1097/qco.0000000000000002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The whole sporozoite (SPZ) vaccine platform provides the only established approach for inducing high-level sustained protective immunity in humans against malaria. We introduce this platform, highlight literature published since 2011, and discuss the challenges of further development. RECENT FINDINGS There are three major approaches to development of a whole parasite vaccine to prevent malaria infection using the SPZ platform: radiation-attenuated sporozoites (irrSPZ), chemoprophylaxis with infectious sporozoites (CPS), and genetically attenuated parasites (GAPs). In all three, SPZ are administered to the vaccinee. All three protect animals against infection when administered by injection with a needle and syringe, and irrSPZ and CPS protect against Plasmodium falciparum malaria in humans when P. falciparum SPZ (PfSPZ) are administered by mosquito bite. Metabolically active, nonreplicating (radiation attenuated) aseptic, purified, cryopreserved PfSPZ (PfSPZ Vaccine), and infectious, aseptic, purified, cryopreserved PfSPZ administered with chemoprophylaxis (PfSPZ-CVac approach) administered by needle and syringe have entered clinical trials. Preliminary data indicate that the PfSPZ Vaccine is safe, well tolerated and highly protective when administered intravenously. SUMMARY With proof-of-concept now established for high-grade protection induced by parenteral administration of a whole sporozoite vaccine, pathways for further development are currently being defined. Demonstration of high-level, durable, cross-strain P. falciparum protection would set the stage for licensure of a vaccine that could lead to dramatic reductions in malaria morbidity and mortality, and eventually elimination of this ancient scourge.
Collapse
|
177
|
Foquet L, Hermsen CC, van Gemert GJ, Van Braeckel E, Weening KE, Sauerwein R, Meuleman P, Leroux-Roels G. Vaccine-induced monoclonal antibodies targeting circumsporozoite protein prevent Plasmodium falciparum infection. J Clin Invest 2014; 124:140-4. [PMID: 24292709 DOI: 10.1172/jci70349] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 09/26/2013] [Indexed: 11/17/2022] Open
Abstract
Malaria, which is the result of Plasmodium falciparum infection, is a global health threat that resulted in 655,000 deaths and 216 million clinical cases in 2010 alone. Recent phase 3 trials with malaria vaccine candidate RTS,S/AS01 (RTS,S) in children has demonstrated modest efficacy against clinical and severe malaria. RTS,S targets the pre-erythrocytic phase of the disease and induces high antibody titers against the P. falciparum circumsporozoite protein (CSP) and a moderate CD4(+) T cell response. The individual contribution of these adaptive immune responses to protection from infection remains unknown. Here, we found that prophylactic administration of anti-CSP mAbs derived from an RTS,S-vaccinated recipient fully protected mice with humanized livers from i.v.- and mosquito bite–delivered P. falciparum sporozoite challenge. Titers of anti-CSP that conveyed full protection were within the range observed in human RTS,S vaccine recipients. Increasing anti-CSP titers resulted in a dose-dependent reduction of the liver parasite burden. These data indicate that RTS,S-induced antibodies are protective and provide sterilizing immunity against P. falciparum infection when reaching or exceeding a critical plasma concentration.
Collapse
|
178
|
Schussek S, Trieu A, Doolan DL. Genome- and proteome-wide screening strategies for antigen discovery and immunogen design. Biotechnol Adv 2014; 32:403-14. [DOI: 10.1016/j.biotechadv.2013.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 11/04/2013] [Accepted: 12/16/2013] [Indexed: 01/17/2023]
|
179
|
Challenges and responses in human vaccine development. Curr Opin Immunol 2014; 28:18-26. [PMID: 24561742 DOI: 10.1016/j.coi.2014.01.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/15/2014] [Accepted: 01/20/2014] [Indexed: 01/01/2023]
Abstract
Human vaccine development remains challenging because of the highly sophisticated evasion mechanisms of pathogens for which vaccines are not yet available. Recent years have witnessed both successes and failures of novel vaccine design and the strength of iterative approaches is increasingly appreciated. These combine discovery of novel antigens, adjuvants and vectors in the preclinical stage with computational analyses of clinical data to accelerate vaccine design. Reverse and structural vaccinology have revealed novel antigen candidates and molecular immunology has led to the formulation of promising adjuvants. Gene expression profiles and immune parameters in patients, vaccinees and healthy controls have formed the basis for biosignatures that will provide guidelines for future vaccine design.
Collapse
|
180
|
Mosha JF, Sturrock HJW, Greenwood B, Sutherland CJ, Gadalla NB, Atwal S, Hemelaar S, Brown JM, Drakeley C, Kibiki G, Bousema T, Chandramohan D, Gosling RD. Hot spot or not: a comparison of spatial statistical methods to predict prospective malaria infections. Malar J 2014; 13:53. [PMID: 24517452 PMCID: PMC3932034 DOI: 10.1186/1475-2875-13-53] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/06/2014] [Indexed: 12/02/2022] Open
Abstract
Background Within affected communities, Plasmodium falciparum infections may be skewed in distribution such that single or small clusters of households consistently harbour a disproportionate number of infected individuals throughout the year. Identifying these hotspots of malaria transmission would permit targeting of interventions and a more rapid reduction in malaria burden across the whole community. This study set out to compare different statistical methods of hotspot detection (SaTScan, kernel smoothing, weighted local prevalence) using different indicators (PCR positivity, AMA-1 and MSP-1 antibodies) for prediction of infection the following year. Methods Two full surveys of four villages in Mwanza, Tanzania were completed over consecutive years, 2010-2011. In both surveys, infection was assessed using nested polymerase chain reaction (nPCR). In addition in 2010, serologic markers (AMA-1 and MSP-119 antibodies) of exposure were assessed. Baseline clustering of infection and serological markers were assessed using three geospatial methods: spatial scan statistics, kernel analysis and weighted local prevalence analysis. Methods were compared in their ability to predict infection in the second year of the study using random effects logistic regression models, and comparisons of the area under the receiver operating curve (AUC) for each model. Sensitivity analysis was conducted to explore the effect of varying radius size for the kernel and weighted local prevalence methods and maximum population size for the spatial scan statistic. Results Guided by AUC values, the kernel method and spatial scan statistics appeared to be more predictive of infection in the following year. Hotspots of PCR-detected infection and seropositivity to AMA-1 were predictive of subsequent infection. For the kernel method, a 1 km window was optimal. Similarly, allowing hotspots to contain up to 50% of the population was a better predictor of infection in the second year using spatial scan statistics than smaller maximum population sizes. Conclusions Clusters of AMA-1 seroprevalence or parasite prevalence that are predictive of infection a year later can be identified using geospatial models. Kernel smoothing using a 1 km window and spatial scan statistics both provided accurate prediction of future infection.
Collapse
Affiliation(s)
- Jacklin F Mosha
- National Institute for Medical Research (NIMR), Mwanza Medical Research Centre, Mwanza, Tanzania.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Abstract
Parasitic diseases caused by helminth and protozoan infections remain one of the largest global public health problems for mankind. While natural immunity in man is rare or slow to develop for many parasites, the immune response is capable of recognizing and responding to infection by utilizing a number of different immunological mechanisms. This special topics journal issue examines many of the key findings in the recent literature regarding the immune response against helminth and protozoan infections, as well as highlighting areas in which our current knowledge falls short. The question of how we can tailor immune responses to prevent or reduce disease burden is a burning question within the field of immunoparasitology.
Collapse
Affiliation(s)
- Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - P'ng Loke
- Department of Microbiology, New York University School of Medicine, New York, NY 10010
| |
Collapse
|
182
|
Villarino N, Schmidt NW. CD8 + T Cell Responses to Plasmodium and Intracellular Parasites. ACTA ACUST UNITED AC 2014; 9:169-178. [PMID: 24741372 PMCID: PMC3983867 DOI: 10.2174/1573395509666131126232327] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 11/14/2013] [Accepted: 11/19/2013] [Indexed: 12/29/2022]
Abstract
Parasitic protozoa are major threats to human health affecting millions of people around the world. Control of these infections by the host immune system relies on a myriad of immunological mechanisms that includes both humoral and cellular immunity. CD8+ T cells contribute to the control of these parasitic infections in both animals and humans. Here, we will focus on the CD8+ T cell response against a subset of these protozoa: Plasmodium, Toxoplasma gondii, Leishmania and Trypanosoma cruzi, with an emphasis on experimental rodent systems. It is evident a complex interaction occurs between CD8+ T cells and the invading protozoa. A detailed understanding of how CD8+ T cells mediate protection should provide the basis for the development of effective vaccines that prevent and control infections by these parasites.
Collapse
Affiliation(s)
- Nicolas Villarino
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Nathan W Schmidt
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
183
|
Successful vaccines for naturally occurring protozoal diseases of animals should guide human vaccine research. A review of protozoal vaccines and their designs. Parasitology 2014; 141:624-40. [PMID: 24476952 PMCID: PMC3961066 DOI: 10.1017/s0031182013002060] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Effective vaccines are available for many protozoal diseases of animals, including vaccines for zoonotic pathogens and for several species of vector-transmitted apicomplexan haemoparasites. In comparison with human diseases, vaccine development for animals has practical advantages such as the ability to perform experiments in the natural host, the option to manufacture some vaccines in vivo, and lower safety requirements. Although it is proper for human vaccines to be held to higher standards, the enduring lack of vaccines for human protozoal diseases is difficult to reconcile with the comparatively immense amount of research funding. Common tactical problems of human protozoal vaccine research include reliance upon adapted rather than natural animal disease models, and an overwhelming emphasis on novel approaches that are usually attempted in replacement of rather than for improvement upon the types of designs used in effective veterinary vaccines. Currently, all effective protozoal vaccines for animals are predicated upon the ability to grow protozoal organisms. Because human protozoal vaccines need to be as effective as animal vaccines, researchers should benefit from a comparison of existing veterinary products and leading experimental vaccine designs. With this in mind, protozoal vaccines are here reviewed.
Collapse
|
184
|
Chen SB, Ju C, Chen JH, Zheng B, Huang F, Xiao N, Zhou X, Ernest T, Zhou XN. Operational research needs toward malaria elimination in China. ADVANCES IN PARASITOLOGY 2014; 86:109-33. [PMID: 25476883 DOI: 10.1016/b978-0-12-800869-0.00005-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Owing to the implementation of a national malaria elimination programme from 2010 to 2020, we performed a systematic review to assess research challenges in the People's Republic of China (P.R. China) and define research priorities in the next few years. A systematic search was conducted for articles published from January 2000 to December 2012 in international journals from PubMed and Chinese journals from the China National Knowledge Infrastructure (CNKI). In total, 2532 articles from CNKI and 308 articles from PubMed published between 2010 and 2012 related to malaria after unrelated references and review or comment were further excluded, and a set of research gaps have been identified that could hinder progress toward malaria elimination in P.R. China. For example, there is a lack of sensitive and specific tests for the diagnosis of malaria cases with low parasitemia, and there is a need for surveillance tools that can evaluate the epidemic status for guiding the elimination strategy. Hence, we argue that malaria elimination will be accelerated in P.R. China through the development of new tests, such as detection of parasite or drug resistance, monitoring glucose-6-phosphate dehydrogenase (G6PD) deficiency, active malaria screening methods, and understanding the effects of the environment and climate variation on vector distribution.
Collapse
Affiliation(s)
- Shen-Bo Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, People's Republic of China
| | - Chuan Ju
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, People's Republic of China
| | - Jun-Hu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, People's Republic of China
| | - Bin Zheng
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, People's Republic of China
| | - Fang Huang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, People's Republic of China
| | - Ning Xiao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, People's Republic of China
| | - Xia Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, People's Republic of China
| | - Tambo Ernest
- Center for Sustainable Malaria Control, Faculty of Natural and Environmental Science; Center for Sustainable Malaria Control, Biochemistry Department, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| | - Xiao-Nong Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, People's Republic of China
| |
Collapse
|
185
|
Model for in vivo assessment of humoral protection against malaria sporozoite challenge by passive transfer of monoclonal antibodies and immune serum. Infect Immun 2013; 82:808-17. [PMID: 24478094 DOI: 10.1128/iai.01249-13] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Evidence from clinical trials of malaria vaccine candidates suggests that both cell-mediated and humoral immunity to pre-erythrocytic parasite stages can provide protection against infection. Novel pre-erythrocytic antibody (Ab) targets could be key to improving vaccine formulations, which are currently based on targeting antigens such as the circumsporozoite protein (CSP). However, methods to assess the effects of sporozoite-specific Abs on pre-erythrocytic infection in vivo remain underdeveloped. Here, we combined passive transfer of monoclonal Abs (MAbs) or immune serum with a luciferase-expressing Plasmodium yoelii sporozoite challenge to assess Ab-mediated inhibition of liver infection in mice. Passive transfer of a P. yoelii CSP MAb showed inhibition of liver infection when mice were challenged with sporozoites either intravenously or by infectious mosquito bite. However, inhibition was most potent for the mosquito bite challenge, leading to a more significant reduction of liver-stage burden and even a lack of progression to blood-stage parasitemia. This suggests that Abs provide effective protection against a natural infection. Inhibition of liver infection was also achieved by passive transfer of immune serum from whole-parasite-immunized mice. Furthermore, we demonstrated that passive transfer of a MAb against P. falciparum CSP inhibited liver-stage infection in a humanized mouse/P. falciparum challenge model. Together, these models constitute unique and sensitive in vivo methods to assess serum-transferable protection against Plasmodium sporozoite challenge.
Collapse
|
186
|
Stanisic DI, Barry AE, Good MF. Escaping the immune system: How the malaria parasite makes vaccine development a challenge. Trends Parasitol 2013; 29:612-22. [DOI: 10.1016/j.pt.2013.10.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/01/2013] [Accepted: 10/01/2013] [Indexed: 10/26/2022]
|
187
|
Abstract
The currently available malaria control tools have allowed malaria elimination in many regions but there remain many regions where malaria control has made little progress. A safe and protective malaria vaccine would be a huge asset for malaria control. Despite the many challenges, efforts continue to design and evaluate malaria vaccine candidates. These candidates target different stages in the life cycle of Plasmodia. The most advanced vaccine candidates target the pre-erythrocytic stages in the life cycle of the parasite and include RTS,S/AS01, which has progressed through clinical development to the stage that it may be licensed in 2015. Attenuated whole-parasite vaccine candidates are highly protective, but there are challenges to manufacture and to administration. Cellular immunity is targeted by the prime-boost approach. Priming vectors trigger only modest responses but these are focused on the recombinant antigen. Boosting vectors trigger strong but broad non-specific responses. The heterologous sequence produces strong immunological responses to the recombinant antigen. Candidates that target the blood stages of the parasite have to result in an immune response that is more effective than the response to an infection to abort or control the infection of merozoites and hence disease. Finally, the sexual stages of the parasite offer another target for vaccine development, which would prevent the transmission of malaria. Today it seems unlikely that any candidate targeting a single antigen will provide complete protection against an organism of the complexity of Plasmodium. A systematic search for vaccine targets and combinations of antigens may be a more promising approach.
Collapse
Affiliation(s)
- Lorenz von Seidlein
- Department of Global health, Menzies School of Health Research, , Casuarina, Northern Territory, Australia
| | | |
Collapse
|
188
|
Park HJ, Guariento M, Maciejewski M, Hauhart R, Tham WH, Cowman AF, Schmidt CQ, Mertens HDT, Liszewski MK, Hourcade DE, Barlow PN, Atkinson JP. Using mutagenesis and structural biology to map the binding site for the Plasmodium falciparum merozoite protein PfRh4 on the human immune adherence receptor. J Biol Chem 2013; 289:450-63. [PMID: 24214979 DOI: 10.1074/jbc.m113.520346] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
To survive and replicate within the human host, malaria parasites must invade erythrocytes. Invasion can be mediated by the P. falciparum reticulocyte-binding homologue protein 4 (PfRh4) on the merozoite surface interacting with complement receptor type 1 (CR1, CD35) on the erythrocyte membrane. The PfRh4 attachment site lies within the three N-terminal complement control protein modules (CCPs 1-3) of CR1, which intriguingly also accommodate binding and regulatory sites for the key complement activation-specific proteolytic products, C3b and C4b. One of these regulatory activities is decay-accelerating activity. Although PfRh4 does not impact C3b/C4b binding, it does inhibit this convertase disassociating capability. Here, we have employed ELISA, co-immunoprecipitation, and surface plasmon resonance to demonstrate that CCP 1 contains all the critical residues for PfRh4 interaction. We fine mapped by homologous substitution mutagenesis the PfRh4-binding site on CCP 1 and visualized it with a solution structure of CCPs 1-3 derived by NMR and small angle x-ray scattering. We cross-validated these results by creating an artificial PfRh4-binding site through substitution of putative PfRh4-interacting residues from CCP 1 into their homologous positions within CCP 8; strikingly, this engineered binding site had an ∼30-fold higher affinity for PfRh4 than the native one in CCP 1. These experiments define a candidate site on CR1 by which P. falciparum merozoites gain access to human erythrocytes in a non-sialic acid-dependent pathway of merozoite invasion.
Collapse
Affiliation(s)
- Hyon Ju Park
- From the Washington University School of Medicine, Division of Rheumatology, Department of Internal Medicine, St. Louis, Missouri 63110
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Abstract
Malaria, which is caused by Plasmodium spp., starts with an asymptomatic phase, during which sporozoites, the parasite form that is injected into the skin by a mosquito, develop into merozoites, the form that infects erythrocytes. This pre-erythrocytic phase is still the most enigmatic in the parasite life cycle, but has long been recognized as an attractive vaccination target. In this Review, we present what has been learned in recent years about the natural history of the pre-erythrocytic stages, mainly using intravital imaging in rodents. We also consider how this new knowledge is in turn changing our understanding of the immune response mounted by the host against the pre-erythrocytic forms.
Collapse
|
190
|
Gómez CE, Perdiguero B, García-Arriaza J, Esteban M. Clinical applications of attenuated MVA poxvirus strain. Expert Rev Vaccines 2013; 12:1395-416. [PMID: 24168097 DOI: 10.1586/14760584.2013.845531] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The highly attenuated poxvirus strain modified vaccinia virus Ankara (MVA) has reached maturity as a vector delivery system and as a vaccine candidate against a broad spectrum of diseases. This has been largely recognized from research on virus-host cell interactions and immunological studies in pre-clinical and clinical trials. This review addresses the studies of MVA vectors used in phase I/II clinical trials, with the aim to provide the main findings obtained on their behavior when tested against relevant human diseases and cancer and also highlights the strategies currently implemented to improve the MVA immunogenicity. The authors assess that MVA vectors are progressing as strong vaccine candidates either alone or when administered in combination with other vectors.
Collapse
Affiliation(s)
- Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | | | | | | |
Collapse
|
191
|
Fowkes FJI, Simpson JA, Beeson JG. Implications of the licensure of a partially efficacious malaria vaccine on evaluating second-generation vaccines. BMC Med 2013; 11:232. [PMID: 24228861 PMCID: PMC4225678 DOI: 10.1186/1741-7015-11-232] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/19/2013] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Malaria is a leading cause of morbidity and mortality, with approximately 225 million clinical episodes and >1.2 million deaths annually attributed to malaria. Development of a highly efficacious malaria vaccine will offer unparalleled possibilities for disease prevention and remains a key priority for long-term malaria control and elimination. DISCUSSION The Malaria Vaccine Technology Roadmap's goal is to 'develop and license a first-generation malaria vaccine that has protective efficacy of more than 50%'. To date, malaria vaccine candidates have only been shown to be partially efficacious (approximately 30% to 60%). However, licensure of a partially effective vaccine will create a number of challenges for the development and progression of new, potentially more efficacious, malaria vaccines in the future. In this opinion piece we discuss the methodological, logistical and ethical issues that may impact on the feasibility and implementation of superiority, non-inferiority and equivalence trials to assess second generation malaria vaccines in the advent of the licensure of a partially efficacious malaria vaccine. CONCLUSIONS Selecting which new malaria vaccines go forward, and defining appropriate methodology for assessment in logistically challenging clinical trials, is crucial. It is imperative that the scientific community considers all the issues and starts planning how second-generation malaria vaccines will advance in the advent of licensure of a partially effective vaccine.
Collapse
Affiliation(s)
- Freya J I Fowkes
- Macfarlane Burnet Institute of Medical Research, 85 Commercial Road, Melbourne, Victoria 3004, Australia.
| | | | | |
Collapse
|
192
|
Bowman NM, Congdon S, Mvalo T, Patel JC, Escamilla V, Emch M, Martinson F, Hoffman I, Meshnick SR, Juliano JJ. Comparative population structure of Plasmodium falciparum circumsporozoite protein NANP repeat lengths in Lilongwe, Malawi. Sci Rep 2013; 3:1990. [PMID: 23771124 PMCID: PMC3683670 DOI: 10.1038/srep01990] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 05/24/2013] [Indexed: 02/05/2023] Open
Abstract
Humoral immunity to Plasmodium falciparum circumsporozoite protein is partly mediated by a polymorphic NANP tetra-amino acid repeat. Antibody response to these repeats is the best correlate of protective immunity to the RTS,S malaria vaccine, but few descriptions of the natural variation of these repeats exist. Using capillary electrophoresis to determine the distribution of NANP repeat size polymorphisms among 98 isolates from Lilongwe, Malawi, we characterised the diversity of P. falciparum infection by several ecological indices. Infection by multiple distinct variants was common, and 20 distinct repeat sizes were identified. Diversity of P. falciparum appeared greater in children (18 variants) than adults (12 variants). There was evidence of genetic distance between different geographic regions by Nei's Standard Genetic Distance, suggesting parasite populations vary locally. We show that P. falciparum is very diverse with respect to NANP repeat length even on a local level and that diversity appears higher in children.
Collapse
Affiliation(s)
- Natalie M Bowman
- University of North Carolina-Chapel Hill, Department of Medicine, Division of Infectious Diseases, Chapel Hill, NC, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Pusic K, Clements D, Kobuch S, Hui G. Antibody and T cell responses in reciprocal prime-boost studies with full-length and truncated merozoite surface protein 1-42 vaccines. PLoS One 2013; 8:e75939. [PMID: 24098747 PMCID: PMC3786974 DOI: 10.1371/journal.pone.0075939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/16/2013] [Indexed: 11/18/2022] Open
Abstract
The P. falciparum Merozoite Surface Protein 1-42 (MSP1-42) is one of the most studied malaria subunit vaccine candidates. The N-terminal fragment of MSP1-42, MSP1-33, is primarily composed of allelic sequences, and has been shown to possess T helper epitopes that influence protective antibody responses toward the C-terminal region, MSP1-19. A truncated MSP1-42 vaccine, Construct 33-I, consisting of exclusively conserved T epitope regions of MSP1-33 expressed in tandem with MSP1-19, was previously shown to be a more effective immunogen than the full-length MSP1-42 vaccine. Here, by way of reciprocal priming/boosting immunization regimens, we studied the immunogenicity of Construct 33-I in the context of recognition by immune responses induced by the full-length native MSP1-42 protein, in order to gauge the effects of pre- and post-exposures to MSP1-42 on vaccine induced responses. Judging by immune responsiveness, antibody and T cell responses, Construct 33-I was effective as the priming antigen followed by full-length MSP1-42 boosting, as well as the boosting antigen following full-length MSP1-42 priming. In particular, Construct 33-I priming elicited the broadest responsiveness in immunized animals subsequently exposed to MSP1-42. Moreover, Construct 33-I, with its conserved MSP1-33 specific T cell epitopes, was equally well recognized by homologous and heterologous allelic forms of MSP1-42. Serum antibodies raised against Construct 33-I efficiently inhibited the growth of parasites carrying the heterologous MSP1-42 allele. These results suggest that Construct 33-I maintains and/or enhances its immunogenicity in an allelic or strain transcending fashion when deployed in populations having prior or subsequent exposures to native MSP1-42s.
Collapse
Affiliation(s)
- Kae Pusic
- University of Hawaii, School of Medicine, Department of Tropical Medicine, Honolulu, Hawaii, United States of America
- * E-mail:
| | - Danielle Clements
- University of Hawaii, School of Medicine, Department of Tropical Medicine, Honolulu, Hawaii, United States of America
| | - Sophie Kobuch
- University of Hawaii, School of Medicine, Department of Tropical Medicine, Honolulu, Hawaii, United States of America
| | - George Hui
- University of Hawaii, School of Medicine, Department of Tropical Medicine, Honolulu, Hawaii, United States of America
| |
Collapse
|
194
|
“Omics” in the study of the major parasitic diseases malaria and schistosomiasis. INFECTION GENETICS AND EVOLUTION 2013; 19:258-73. [DOI: 10.1016/j.meegid.2013.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 07/04/2013] [Accepted: 07/07/2013] [Indexed: 01/21/2023]
|
195
|
Warimwe GM, Fletcher HA, Olotu A, Agnandji ST, Hill AVS, Marsh K, Bejon P. Peripheral blood monocyte-to-lymphocyte ratio at study enrollment predicts efficacy of the RTS,S malaria vaccine: analysis of pooled phase II clinical trial data. BMC Med 2013; 11:184. [PMID: 23962071 PMCID: PMC3765422 DOI: 10.1186/1741-7015-11-184] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/26/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND RTS,S is the most advanced candidate malaria vaccine but it is only partially protective and the causes of inter-individual variation in efficacy are poorly understood. Here, we investigated whether peripheral blood monocyte-to-lymphocyte ratios (ML ratio), previously shown to correlate with clinical malaria risk, could account for differences in RTS,S efficacy among phase II trial participants in Africa. METHODS Of 11 geographical sites where RTS,S has been evaluated, pre-vaccination ML ratios were only available for trial participants in Kilifi, Kenya (N = 421) and Lambarene, Gabon (N = 189). Using time to first clinical malaria episode as the primary endpoint we evaluated the effect of accounting for ML ratio on RTS,S vaccine efficacy against clinical malaria by Cox regression modeling. RESULTS The unadjusted efficacy of RTS,S in this combined dataset was 47% (95% confidence interval (CI) 26% to 62%, P <0.001). However, RTS,S efficacy decreased with increasing ML ratio, ranging from 67% (95% CI 64% to 70%) at an ML ratio of 0.1 to 5% (95% CI -3% to 13%) at an ML ratio of 0.6. The statistical interaction between RTS,S vaccination and ML ratio was still evident after adjustment for covariates associated with clinical malaria risk in this dataset. CONCLUSION The results suggest that stratification of study participants by ML ratio, easily measured from full differential blood counts before vaccination, might help identify children who are highly protected and those that are refractory to protection with the RTS,S vaccine. Identifying causes of low vaccine efficacy among individuals with high ML ratio could inform strategies to improve overall RTS,S vaccine efficacy. TRIAL REGISTRATION ClinicalTrials.Gov numbers NCT00380393 and NCT00436007.
Collapse
Affiliation(s)
- George M Warimwe
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
196
|
Rosenthal PJ. The interplay between drug resistance and fitness in malaria parasites. Mol Microbiol 2013; 89:1025-38. [PMID: 23899091 DOI: 10.1111/mmi.12349] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2013] [Indexed: 12/01/2022]
Abstract
Controlling the spread of antimalarial drug resistance, especially resistance of Plasmodium falciparum to artemisinin-based combination therapies, is a high priority. Available data indicate that, as with other microorganisms, the spread of drug-resistant malaria parasites is limited by fitness costs that frequently accompany resistance. Resistance-mediating polymorphisms in malaria parasites have been identified in putative drug transporters and in target enzymes. The impacts of these polymorphisms on parasite fitness have been characterized in vitro and in animal models. Additional insights have come from analyses of samples from clinical studies, both evaluating parasites under different selective pressures and determining the clinical consequences of infection with different parasites. With some exceptions, resistance-mediating polymorphisms lead to malaria parasites that, compared with wild type, grow less well in culture and in animals, and are replaced by wild type when drug pressure diminishes in the clinical setting. In some cases, the fitness costs of resistance may be offset by compensatory mutations that increase virulence or changes that enhance malaria transmission. However, not enough is known about effects of resistance mediators on parasite fitness. A better appreciation of the costs of fitness-mediating mutations will facilitate the development of optimal guidelines for the treatment and prevention of malaria.
Collapse
Affiliation(s)
- Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
197
|
Abstract
PURPOSE OF REVIEW Vector-borne diseases (VBDs) are difficult to prevent and control because it is hard to predict the complex habits of mosquitoes, ticks and fleas; most vector-borne viruses or bacteria infect animals as well as humans, which further adds to this difficulty. Thus, prevention is the best protection against VBD. RECENT FINDINGS Vaccines are available for yellow fever, Japanese encephalitis and tick-borne encephalitis and several vaccines are in clinical trials for dengue fever. Antimalarial intermittent preventive therapy (sulfadoxine-pyrimethamine) and insecticide-treated mosquito nets are associated with a decreased risk of neonatal mortality and lower birth-weight. Permethrin-impregnated clothing for the prevention of tick bites has been shown effective in reducing tick bites. SUMMARY Much progress has been made in terms of development of preventive vaccines and medicines, but there is more work that needs to be done. Efforts still need to continue on raising awareness for prevention of VBD.
Collapse
|
198
|
Immunogenicity of recombinant proteins consisting of Plasmodium vivax circumsporozoite protein allelic variant-derived epitopes fused with Salmonella enterica Serovar Typhimurium flagellin. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:1418-25. [PMID: 23863502 DOI: 10.1128/cvi.00312-13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
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.
Collapse
|
199
|
Frech C, Chen N. Variant surface antigens of malaria parasites: functional and evolutionary insights from comparative gene family classification and analysis. BMC Genomics 2013; 14:427. [PMID: 23805789 PMCID: PMC3747859 DOI: 10.1186/1471-2164-14-427] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 06/19/2013] [Indexed: 11/10/2022] Open
Abstract
Background Plasmodium parasites, the causative agents of malaria, express many variant antigens on cell surfaces. Variant surface antigens (VSAs) are typically organized into large subtelomeric gene families that play critical roles in virulence and immune evasion. Many important aspects of VSA function and evolution remain obscure, impeding our understanding of virulence mechanisms and vaccine development. To gain further insights into VSA function and evolution, we comparatively classified and examined known VSA gene families across seven Plasmodium species. Results We identified a set of ultra-conserved orthologs within the largest Plasmodium gene family pir, which should be considered as high-priority targets for experimental functional characterization and vaccine development. Furthermore, we predict a lipid-binding domain in erythrocyte surface-expressed PYST-A proteins, suggesting a role of this second largest rodent parasite gene family in host cholesterol salvage. Additionally, it was found that PfMC-2TM proteins carry a novel and putative functional domain named MC-TYR, which is conserved in other P. falciparum gene families and rodent parasites. Finally, we present new conclusive evidence that the major Plasmodium VSAs PfEMP1, SICAvar, and SURFIN are evolutionarily linked through a modular and structurally conserved intracellular domain. Conclusion Our comparative analysis of Plasmodium VSA gene families revealed important functional and evolutionary insights, which can now serve as starting points for further experimental studies.
Collapse
Affiliation(s)
- Christian Frech
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | | |
Collapse
|
200
|
Febir LG, Asante KP, Dzorgbo DBS, Senah KA, Letsa TS, Owusu-Agyei S. Community perceptions of a malaria vaccine in the Kintampo districts of Ghana. Malar J 2013; 12:156. [PMID: 23651533 PMCID: PMC3656774 DOI: 10.1186/1475-2875-12-156] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 05/06/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Malaria remains the leading cause of morbidity and mortality in sub-Saharan Africa despite tools currently available for its control. Making malaria vaccine available for routine use will be a major hallmark, but its acceptance by community members and health professionals within the health system could pose considerable challenge as has been found with the introduction of polio vaccinations in parts of West Africa. Some of these challenges may not be expected since decisions people make are many a time driven by a complex myriad of perceptions. This paper reports knowledge and perceptions of community members in the Kintampo area of Ghana where malaria vaccine trials have been ongoing as part of the drive for the first-ever licensed malaria vaccine in the near future. METHODS Both qualitative and quantitative methods were used in the data collection processes. Women and men whose children were or were not involved in the malaria vaccine trial were invited to participate in focus group discussions (FGDs). Respondents, made up of heads of religious groupings in the study area, health care providers, traditional healers and traditional birth attendants, were also invited to participate in in-depth interviews (IDIs). A cross-sectional survey was conducted in communities where the malaria vaccine trial (Mal 047RTS,S) was carried out. In total, 12 FGDs, 15 IDIs and 466 household head interviews were conducted. RESULTS Knowledge about vaccines was widespread among participants. Respondents would like their children to be vaccinated against all childhood illnesses including malaria. Knowledge of the long existing routine vaccines was relatively high among respondents compared to hepatitis B and Haemophilus influenza type B vaccines that were introduced more recently in 2002. There was no clear religious belief or sociocultural practice that will serve as a possible barrier to the acceptance of a malaria vaccine. CONCLUSION With the assumption that a malaria vaccine will be as efficacious as other EPI vaccines, community members in Central Ghana will accept and prefer malaria vaccine to malaria drugs as a malaria control tool. Beliefs and cultural practices as barriers to the acceptance of malaria vaccine were virtually unknown in the communities surveyed.
Collapse
|