151
|
Yap XZ, McCall MBB, Sauerwein RW. Fast and fierce versus slow and smooth: Heterogeneity in immune responses to Plasmodium in the controlled human malaria infection model. Immunol Rev 2020; 293:253-269. [PMID: 31605396 PMCID: PMC6973142 DOI: 10.1111/imr.12811] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022]
Abstract
Controlled human malaria infection (CHMI) is an established model in clinical malaria research. Upon exposure to Plasmodium falciparum parasites, malaria-naive volunteers differ in dynamics and composition of their immune profiles and subsequent capacity to generate protective immunity. CHMI volunteers are either inflammatory responders who have prominent cellular IFN-γ production primarily driven by adaptive T cells, or tempered responders who skew toward antibody-mediated humoral immunity. When exposed to consecutive CHMIs under antimalarial chemoprophylaxis, individuals who can control parasitemia after a single immunization (fast responders) are more likely to be protected against a subsequent challenge infection. Fast responders tend to be inflammatory responders who can rapidly induce long-lived IFN-γ+ T cell responses. Slow responders or even non-responders can also be protected, but via a more diverse range of responses that take a longer time to reach full protective efficacy, in part due to their tempered phenotype. The latter group can be identified at baseline before CHMI by higher expression of inhibitory ligands CTLA-4 and TIM-3 on CD4+ T cells. Delineating heterogeneity in human immune responses to P. falciparum will facilitate rational design and strategy towards effective malaria vaccines.
Collapse
Affiliation(s)
- Xi Zen Yap
- Department of Medical MicrobiologyRadboud University Medical CenterNijmegenThe Netherlands
- Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
| | - Matthew B. B. McCall
- Department of Medical MicrobiologyRadboud University Medical CenterNijmegenThe Netherlands
- Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
| | - Robert W. Sauerwein
- Department of Medical MicrobiologyRadboud University Medical CenterNijmegenThe Netherlands
- Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
152
|
[Development of malaria vaccines-state of the art]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2019; 63:45-55. [PMID: 31828371 PMCID: PMC7223738 DOI: 10.1007/s00103-019-03070-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Weltweit leben 3,1 Mrd. Menschen in Gebieten, in denen Malaria endemisch ist (Tropen, Subtropen). Jährlich erkranken etwa 200 Mio. Menschen, ca. 500.000 sterben daran. Betroffen sind vor allem Kinder. Um die Malaria zu kontrollieren und schlussendlich jegliche Neuinfektion zu verhindern, ist die Entwicklung wirksamer Impfstoffe von großer Bedeutung. In diesem Beitrag werden zunächst Hintergrundinformationen zur Geschichte der Impfstoffentwicklung, zur Malariaerkrankung und zu den Möglichkeiten der Therapie und Ausbreitungskontrolle gegeben. Der Hauptteil widmet sich dem aktuellen Forschungsstand zu Impfstoffen gegen den Erreger Plasmodium falciparum, gefolgt von einer ausführlichen Diskussion. Malaria ist eine parasitäre Infektionskrankheit, die von Einzellern, sog. Plasmodien, verursacht wird. Es werden 5 humanpathogene Spezies unterschieden, von denen P. falciparum über 99 % der Erkrankungen in Afrika verursacht. Überträger (Vektor) ist die Anophelesmücke. Plasmodium bietet innerhalb seines Lebenszyklus verschiedene Ansatzpunkte für die Wirkung von Impfstoffen. Von den insgesamt ca. 70 Impfstoffkandidaten sind die präerythrozytären Impfstoffe, die in den Leberzyklus des Parasiten eingreifen, aktuell am weitesten entwickelt. Die von der Weltgesundheitsorganisation (WHO) angestrebte Wirksamkeit von mindestens 75 % wurde aber längst nicht erreicht. Mit RTS,S/AS01 wird derzeit erstmals ein mäßig wirksamer Impfstoff großflächig eingesetzt. Schon jetzt ist offensichtlich, dass die Malaria nur im Zusammenspiel mit anderen Maßnahmen eingedämmt werden kann. Expositionsprophylaxe mit imprägnierten Moskitonetzen, der Einsatz von Insektiziden mit Residualeffekt in Innenräumen (Indoor Residual Spraying), die Vernichtung von Moskitobrutplätzen und schnelle Diagnose und Therapie der Erkrankung sind hier wichtige Elemente ebenso wie eine funktionierende Gesundheitsversorgung, die in den von Armut geprägten Gebieten oft nicht gewährleistet ist.
Collapse
|
153
|
Rutishauser T, Lepore M, Di Blasi D, Dangy JP, Abdulla S, Jongo S, Ramadhani K, Sim BKL, Hoffman SL, Tanner M, Daubenberger C, De Libero G. Activation of TCR Vδ1 + and Vδ1 -Vδ2 - γδ T Cells upon Controlled Infection with Plasmodium falciparum in Tanzanian Volunteers. THE JOURNAL OF IMMUNOLOGY 2019; 204:180-191. [PMID: 31801816 DOI: 10.4049/jimmunol.1900669] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/04/2019] [Indexed: 01/05/2023]
Abstract
Our understanding of the human immune response to malaria remains incomplete. Clinical trials using whole-sporozoite-based vaccination approaches such as the Sanaria PfSPZ Vaccine, followed by controlled human malaria infection (CHMI) to assess vaccine efficacy offer a unique opportunity to study the immune response during Plasmodium falciparum infection. Diverse populations of T cells that are not restricted to classical HLA (unconventional T cells) participate in the host response during Plasmodium infection. Although several populations of unconventional T cells exist, the majority of studies focused on TCR Vγ9Vδ2 cells, the most abundant TCR γδ cell population in peripheral blood. In this study, we dissected the response of three TCR γδ cell subsets and mucosal-associated invariant T cells in healthy volunteers immunized with PfSPZ Vaccine and challenged by CHMI using Sanaria PfSPZ Challenge. Using a flow cytometry-based unbiased analysis followed by T cell cloning, several findings were made. Whereas major ex vivo alterations were not detectable after immunization with PfSPZ Vaccine, TCR Vδ2, and mucosal-associated invariant T cells expanded after asexual blood-stage parasitemia induced by CHMI. CHMI, but not vaccination, also induced the activation of TCR Vδ1 and Vδ1-Vδ2- γδ T cells. The activated TCR Vδ1 cells were oligoclonal, suggesting clonal expansion, and upon repeated CHMI, showed diminished response, indicating long-term alterations induced by blood-stage parasitemia. Some TCR Vδ1 clones recognized target cells in the absence of parasite-derived Ags, thus suggesting recognition of self-molecules. These findings reveal the articulate participation of different populations of unconventional T cells to P. falciparum infection.
Collapse
Affiliation(s)
- Tobias Rutishauser
- Clinical Immunology Unit, Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland.,University of Basel, 4001 Basel, Switzerland.,Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Marco Lepore
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Daniela Di Blasi
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Jean-Pierre Dangy
- Clinical Immunology Unit, Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland.,University of Basel, 4001 Basel, Switzerland
| | - Salim Abdulla
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania; and
| | - Said Jongo
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania; and
| | - Kamaka Ramadhani
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania; and
| | | | | | - Marcel Tanner
- Clinical Immunology Unit, Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland.,University of Basel, 4001 Basel, Switzerland
| | - Claudia Daubenberger
- Clinical Immunology Unit, Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland; .,University of Basel, 4001 Basel, Switzerland
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland;
| |
Collapse
|
154
|
Kapulu MC, Njuguna P, Hamaluba MM. Controlled Human Malaria Infection in Semi-Immune Kenyan Adults (CHMI-SIKA): a study protocol to investigate in vivo Plasmodium falciparum malaria parasite growth in the context of pre-existing immunity. Wellcome Open Res 2019; 3:155. [PMID: 31803847 PMCID: PMC6871356 DOI: 10.12688/wellcomeopenres.14909.2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2019] [Indexed: 01/09/2023] Open
Abstract
Malaria remains a major public health burden despite approval for implementation of a partially effective pre-erythrocytic malaria vaccine. There is an urgent need to accelerate development of a more effective multi-stage vaccine. Adults in malaria endemic areas may have substantial immunity provided by responses to the blood stages of malaria parasites, but field trials conducted on several blood-stage vaccines have not shown high levels of efficacy. We will use the controlled human malaria infection (CHMI) models with malaria-exposed volunteers to identify correlations between immune responses and parasite growth rates in vivo. Immune responses more strongly associated with control of parasite growth should be prioritized to accelerate malaria vaccine development. We aim to recruit up to 200 healthy adult volunteers from areas of differing malaria transmission in Kenya, and after confirming their health status through clinical examination and routine haematology and biochemistry, we will comprehensively characterize immunity to malaria using >100 blood-stage antigens. We will administer 3,200 aseptic, purified, cryopreserved Plasmodium falciparum sporozoites (PfSPZ Challenge) by direct venous inoculation. Serial quantitative polymerase chain reaction to measure parasite growth rate in vivo will be undertaken. Clinical and laboratory monitoring will be undertaken to ensure volunteer safety. In addition, we will also explore the perceptions and experiences of volunteers and other stakeholders in participating in a malaria volunteer infection study. Serum, plasma, peripheral blood mononuclear cells and whole blood will be stored to allow a comprehensive assessment of adaptive and innate host immunity. We will use CHMI in semi-immune adult volunteers to relate parasite growth outcomes with antibody responses and other markers of host immunity. Registration: ClinicalTrials.gov identifier NCT02739763.
Collapse
Affiliation(s)
- Melissa C. Kapulu
- KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, UK
| | | | | | - CHMI-SIKA Study Team
- KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, UK
| |
Collapse
|
155
|
Good MF, Stanisic DI. Whole parasite vaccines for the asexual blood stages ofPlasmodium. Immunol Rev 2019; 293:270-282. [DOI: 10.1111/imr.12819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Michael F. Good
- Institute for Glycomics Griffith University Gold Coast Qld. Australia
| | | |
Collapse
|
156
|
Prospects for Malaria Vaccines: Pre-Erythrocytic Stages, Blood Stages, and Transmission-Blocking Stages. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9751471. [PMID: 31687404 PMCID: PMC6794966 DOI: 10.1155/2019/9751471] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/06/2019] [Accepted: 09/09/2019] [Indexed: 12/18/2022]
Abstract
Malaria is a disease of public health importance in many parts of the world. Currently, there is no effective way to eradicate malaria, so developing safe, efficient, and cost-effective vaccines against this disease remains an important goal. Current research on malaria vaccines is focused on developing vaccines against pre-erythrocytic stage parasites and blood-stage parasites or on developing a transmission-blocking vaccine. Here, we briefly describe the progress made towards a vaccine against Plasmodium falciparum, the most pathogenic of the malaria parasite species to infect humans.
Collapse
|
157
|
Goswami D, Minkah NK, Kappe SHI. Designer Parasites: Genetically Engineered Plasmodium as Vaccines To Prevent Malaria Infection. THE JOURNAL OF IMMUNOLOGY 2019; 202:20-28. [PMID: 30587570 DOI: 10.4049/jimmunol.1800727] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/21/2018] [Indexed: 12/20/2022]
Abstract
A highly efficacious malaria vaccine that prevents disease and breaks the cycle of infection remains an aspirational goal of medicine. Whole parasite vaccines based on the sporozoite forms of the parasite that target the clinically silent pre-erythrocytic stages of infection have emerged as one of the leading candidates. In animal models of malaria, these vaccines elicit potent neutralizing Ab responses against the sporozoite stage and cytotoxic T cells that eliminate parasite-infected hepatocytes. Among whole-sporozoite vaccines, immunization with live, replication-competent whole parasites engenders superior immunity and protection when compared with live replication-deficient sporozoites. As such, the genetic design of replication-competent vaccine strains holds the promise for a potent, broadly protective malaria vaccine. In this report, we will review the advances in whole-sporozoite vaccine development with a particular focus on genetically attenuated parasites both as malaria vaccine candidates and also as valuable tools to interrogate protective immunity against Plasmodium infection.
Collapse
Affiliation(s)
- Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109; and
| | - Nana K Minkah
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109; and
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109; and .,Department of Global Health, University of Washington, Seattle, WA 98195
| |
Collapse
|
158
|
Tran TM, Crompton PD. Decoding the complexities of human malaria through systems immunology. Immunol Rev 2019; 293:144-162. [PMID: 31680289 DOI: 10.1111/imr.12817] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
The complexity of the Plasmodium parasite and its life cycle poses a challenge to our understanding of the host immune response against malaria. Studying human immune responses during natural and experimental Plasmodium infections can enhance our understanding of malaria-protective immunity and inform the design of disease-modifying adjunctive therapies and next-generation malaria vaccines. Systems immunology can complement conventional approaches to facilitate our understanding of the complex immune response to the highly dynamic malaria parasite. In this review, recent studies that used systems-based approaches to evaluate human immune responses during natural and experimental Plasmodium falciparum and Plasmodium vivax infections as well as during immunization with candidate malaria vaccines are summarized and related to each other. The potential for next-generation technologies to address the current limitations of systems-based studies of human malaria are discussed.
Collapse
Affiliation(s)
- Tuan M Tran
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Ryan White Center for Pediatric Infectious Disease and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
159
|
Kumar R, Loughland JR, Ng SS, Boyle MJ, Engwerda CR. The regulation of CD4
+
T cells during malaria. Immunol Rev 2019; 293:70-87. [DOI: 10.1111/imr.12804] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Rajiv Kumar
- Centre of Experimental Medicine and Surgery Institute of Medical Sciences Banaras Hindu University Varanasi UP India
- Department of Medicine Institute of Medical Sciences Banaras Hindu University Varanasi UP India
| | - Jessica R. Loughland
- Human Malaria Immunology Laboratory QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Susanna S. Ng
- Immunology and Infection Laboratory QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Michelle J. Boyle
- Human Malaria Immunology Laboratory QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Christian R. Engwerda
- Immunology and Infection Laboratory QIMR Berghofer Medical Research Institute Brisbane Australia
| |
Collapse
|
160
|
Narula AK, Azad CS, Nainwal LM. New dimensions in the field of antimalarial research against malaria resurgence. Eur J Med Chem 2019; 181:111353. [DOI: 10.1016/j.ejmech.2019.05.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/16/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022]
|
161
|
Abstract
Malaria is an illness caused by Plasmodium parasites transmitted to humans by infected mosquitoes. Of the five species that infect humans, P. falciparum exacts the highest toll in terms of human morbidity and mortality, and therefore represents a major public health threat in endemic areas. Recent advances in control efforts have reduced malaria incidence and prevalence, including rapid diagnostic testing, highly effective artemisinin combination therapy, use of insecticide-treated bednets, and indoor residual spraying. But, reductions in numbers of cases have stalled over the last few years, and incidence may have increased. As this concerning trend calls for new tools to combat the disease, the RTS,S vaccine has arrived just in time. The vaccine was created in 1987 and began pilot implementation in endemic countries in 2019. This first-generation malaria vaccine demonstrates modest efficacy against malaria illness and holds promise as a public health tool, especially for children in high-transmission areas where mortality is high.
Collapse
Affiliation(s)
- Matthew B Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
162
|
Hoo R, Bruske E, Dimonte S, Zhu L, Mordmüller B, Sim BKL, Kremsner PG, Hoffman SL, Bozdech Z, Frank M, Preiser PR. Transcriptome profiling reveals functional variation in Plasmodium falciparum parasites from controlled human malaria infection studies. EBioMedicine 2019; 48:442-452. [PMID: 31521613 PMCID: PMC6838377 DOI: 10.1016/j.ebiom.2019.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/29/2019] [Accepted: 09/01/2019] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The transcriptome of Plasmodium falciparum clinical isolates varies according to strain, mosquito bites, disease severity and clinical history. Therefore, it remains a challenge to directly interpret the parasite's transcriptomic information into a more general biological signature in a natural human malaria infection. These confounding variations can be potentially overcome with parasites derived from controlled-human malaria infection (CHMI) studies. METHODS We performed CHMI studies in healthy and immunologically naïve volunteers receiving the same P. falciparum strain ((Sanaria® PfSPZ Challenge (NF54)), but with different sporozoite dosage and route of infection. Parasites isolated from these volunteers at the day of patency were subjected to in vitro culture for several generations and synchronized ring-stage parasites were subjected to transcriptome profiling. FINDINGS We observed clear deviations between CHMI-derived parasites from volunteer groups receiving different PfSPZ dose and route. CHMI-derived parasites and the pre-mosquito strain used for PfSPZ generation showed significant transcriptional variability for gene clusters associated with malaria pathogenesis, immune evasion and transmission. These transcriptional variation signature clusters were also observed in the transcriptome of P. falciparum isolates from acute clinical infections. INTERPRETATION Our work identifies a previously unrecognized transcriptional pattern in malaria infections in a non-immune background. Significant transcriptome heterogeneity exits between parasites derived from human infections and the pre-mosquito strain, implying that the malaria parasites undergo a change in functional state to adapt to its host environment. Our work also highlights the potential use of transcriptomics data from CHMI study advance our understanding of malaria parasite adaptation and transmission in humans. FUND: This work is supported by German Israeli Foundation, German ministry for education and research, MOE Tier 1 from the Singapore Ministry of Education Academic Research Fund, Singapore Ministry of Health's National Medical Research Council, National Institute of Allergy and Infectious Diseases, National Institutes of Health, USA and the German Centre for Infection Research (Deutsches Zentrum für Infektionsforschung-DZIF).
Collapse
Affiliation(s)
- Regina Hoo
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Ellen Bruske
- Institute of Tropical Medicine, Wilhelmstr. 27, University of Tübingen, 72074 Tübingen, Germany
| | - Sandra Dimonte
- Institute of Tropical Medicine, Wilhelmstr. 27, University of Tübingen, 72074 Tübingen, Germany
| | - Lei Zhu
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Benjamin Mordmüller
- Institute of Tropical Medicine, Wilhelmstr. 27, University of Tübingen, 72074 Tübingen, Germany; German Center for Infection Research, partner site Tübingen, Germany
| | - B Kim Lee Sim
- Sanaria Inc, 9800 Medical Center Dr A209, Rockville, MD 20850, USA
| | - Peter G Kremsner
- Institute of Tropical Medicine, Wilhelmstr. 27, University of Tübingen, 72074 Tübingen, Germany; Centre de Recherches Médicales de Lambaréné, BP 242 Lambaréné, Gabon
| | | | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Matthias Frank
- Institute of Tropical Medicine, Wilhelmstr. 27, University of Tübingen, 72074 Tübingen, Germany.
| | - Peter R Preiser
- School of Biological Sciences, Nanyang Technological University, Singapore.
| |
Collapse
|
163
|
Dejon-Agobe JC, Ateba-Ngoa U, Lalremruata A, Homoet A, Engelhorn J, Nouatin OP, Edoa JR, Fernandes JF, Esen M, Mouwenda YD, Betouke Ongwe EM, Massinga-Loembe M, Hoffman SL, Sim BKL, Theisen M, Kremsner PG, Adegnika AA, Lell B, Mordmüller B. Controlled Human Malaria Infection of Healthy Adults With Lifelong Malaria Exposure to Assess Safety, Immunogenicity, and Efficacy of the Asexual Blood Stage Malaria Vaccine Candidate GMZ2. Clin Infect Dis 2019; 69:1377-1384. [PMID: 30561539 PMCID: PMC6763635 DOI: 10.1093/cid/ciy1087] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/18/2018] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND GMZ2 is a recombinant malaria vaccine inducing immune responses against Plasmodium falciparum (Pf) merozoite surface protein-3 and glutamate-rich protein. We used standardized controlled human malaria infection (CHMI) to assess the efficacy of this asexual blood-stage vaccine. METHODS We vaccinated 50 healthy, adult volunteers with lifelong exposure to Pf 3 times, at 4-week intervals, with 30 or 100 µg GMZ2 formulated in CAF01, a liposome-based adjuvant; 100 µg GMZ2, formulated in Alhydrogel; or a control vaccine (Verorab). Approximately 13 weeks after the last vaccination, 35/50 volunteers underwent CHMI by direct venous inoculation of 3200 Pf sporozoites (Sanaria® PfSPZ Challenge). RESULTS Adverse events were similarly distributed between GMZ2 and control vaccinees. Baseline-corrected anti-GMZ2 antibody concentrations 4 weeks after the last vaccination were higher in all 3 GMZ2-vaccinated arms, compared to the control group. All GMZ2 formulations induced similar antibody levels. CHMI resulted in 29/34 (85%) volunteers with Pf parasitemia and 15/34 (44%) with malaria (parasitemia and symptoms). The proportion of participants with malaria (2/5 control, 6/10 GMZ2-Alhydrogel, 2/8 30 µg GMZ2-CAF01, and 5/11 100 µg GMZ2-CAF01) and the time it took them to develop malaria were similar in all groups. Baseline, vaccine-specific antibody concentrations were associated with protection against malaria. CONCLUSIONS GMZ2 is well tolerated and immunogenic in lifelong-Pf-exposed adults from Gabon, with similar antibody responses regardless of formulation. CHMI showed no protective effect of prior vaccination with GMZ2, although baseline, vaccine-specific antibody concentrations were associated with protection. CHMI with the PfSPZ Challenge is a potent new tool to validate asexual, blood-stage malaria vaccines in Africa. CLINICAL TRIALS REGISTRATION Pan-African Clinical Trials: PACTR201503001038304.
Collapse
Affiliation(s)
- Jean Claude Dejon-Agobe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Ulysse Ateba-Ngoa
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Albert Lalremruata
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Andreas Homoet
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Julie Engelhorn
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Odilon Paterne Nouatin
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Jean Ronald Edoa
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - José F Fernandes
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Meral Esen
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Yoanne Darelle Mouwenda
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Eunice M Betouke Ongwe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Marguerite Massinga-Loembe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | | | | | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Denmark
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Ayôla A Adegnika
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| |
Collapse
|
164
|
Zenklusen I, Jongo S, Abdulla S, Ramadhani K, Lee Sim BK, Cardamone H, Flannery EL, Nguyen T, Fishbaugher M, Steel RWJ, Betz W, Carmago N, Mikolajczak S, Kappe SHI, Hoffman SL, Sack BK, Daubenberger C. Immunization of Malaria-Preexposed Volunteers With PfSPZ Vaccine Elicits Long-Lived IgM Invasion-Inhibitory and Complement-Fixing Antibodies. J Infect Dis 2019; 217:1569-1578. [PMID: 29438525 DOI: 10.1093/infdis/jiy080] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/06/2018] [Indexed: 01/17/2023] Open
Abstract
Background The assessment of antibody responses after immunization with radiation-attenuated, aseptic, purified, cryopreserved Plasmodium falciparum sporozoites (Sanaria PfSPZ Vaccine) has focused on IgG isotype antibodies. Here, we aimed to investigate if P. falciparum sporozoite binding and invasion-inhibitory IgM antibodies are induced following immunization of malaria-preexposed volunteers with PfSPZ Vaccine. Methods Using serum from volunteers immunized with PfSPZ, we measured vaccine-induced IgG and IgM antibodies to P. falciparum circumsporozoite protein (PfCSP) via ELISA. Function of this serum as well as IgM antibody fractions was measured via in vitro in an inhibition of sporozoite invasion assay. These IgM antibody fractions were also measured for binding to sporozoites by immunofluorescence assay and complement fixation on whole sporozoites. Results We found that in addition to anti-PfCSP IgG, malaria-preexposed volunteers developed anti-PfCSP IgM antibodies after immunization with PfSPZ Vaccine and that these IgM antibodies inhibited P. falciparum sporozoite invasion of hepatocytes in vitro. These IgM plasma fractions also fixed complement to whole P. falciparum sporozoites. Conclusions This is the first finding that PfCSP and P. falciparum sporozoite-binding IgM antibodies are induced following immunization of PfSPZ Vaccine in malaria-preexposed individuals and that IgM antibodies can inhibit P. falciparum sporozoite invasion into hepatocytes in vitro and fix complement on sporozoites. These findings indicate that the immunological assessment of PfSPZ Vaccine-induced antibody responses could be more sensitive if they include parasite-specific IgM in addition to IgG antibodies. Clinical Trials Registration NCT02132299.
Collapse
Affiliation(s)
- Isabelle Zenklusen
- Clinical Immunology Unit, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| | - Said Jongo
- Ifakara Health Institute, Clinical Trial Unit, Bagamoyo, Tanzania
| | - Salim Abdulla
- Ifakara Health Institute, Clinical Trial Unit, Bagamoyo, Tanzania
| | - Kamaka Ramadhani
- Ifakara Health Institute, Clinical Trial Unit, Bagamoyo, Tanzania
| | | | | | | | - Thao Nguyen
- Center for Infectious Disease Research, Seattle, Washington
| | | | - Ryan W J Steel
- Center for Infectious Disease Research, Seattle, Washington
| | - Will Betz
- Center for Infectious Disease Research, Seattle, Washington
| | - Nelly Carmago
- Center for Infectious Disease Research, Seattle, Washington
| | | | - Stefan H I Kappe
- Center for Infectious Disease Research, Seattle, Washington.,Department of Global Health, University of Washington, Seattle
| | | | - Brandon K Sack
- Center for Infectious Disease Research, Seattle, Washington
| | - Claudia Daubenberger
- Clinical Immunology Unit, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Switzerland
| |
Collapse
|
165
|
Mo AX, McGugan G. Understanding the Liver-Stage Biology of Malaria Parasites: Insights to Enable and Accelerate the Development of a Highly Efficacious Vaccine. Am J Trop Med Hyg 2019; 99:827-832. [PMID: 30141395 PMCID: PMC6159572 DOI: 10.4269/ajtmh.17-0895] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
In August 2017, the National Institute of Allergy and Infectious Diseases convened a meeting, entitled "Understanding the Liver-Stage Biology of Malaria Parasites to Enable and Accelerate the Development of a Highly Efficacious Vaccine," to discuss the needs and strategies to develop a highly efficacious, whole organism-based vaccine targeting the liver stage of malaria parasites. It was concluded that attenuated sporozoite platforms have proven to be promising approaches, and that late-arresting sporozoites could potentially offer greater vaccine performance than early-arresting sporozoites against malaria. New knowledge and emerging technologies have made the development of late-arresting sporozoites feasible. Highly integrated approaches involving liver-stage research, "omics" studies, and cutting-edge genetic editing technologies, combined with in vitro culture systems or unique animal models, are needed to accelerate the discovery of candidates for a late-arresting, genetically attenuated parasite vaccine.
Collapse
Affiliation(s)
- Annie X Mo
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Maryland
| | - Glen McGugan
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Maryland
| |
Collapse
|
166
|
Abstract
Malaria vaccine development has rapidly advanced in the past decade. The very first phase 3 clinical trial of the RTS,S vaccine was completed with over 15,000 African infants and children, and pilot implementation studies are underway. Next-generation candidate vaccines using novel antigens, platforms, or approaches targeting different and/or multiple stages of the Plasmodium life cycle are being tested. Many candidates, in various stages of development, promise enhanced efficacy of long duration and broad protection against genetically diverse malaria strains, with a few studies under way in target populations in endemic areas. Malaria vaccines together with other interventions promise interruption and eventual elimination of malaria in endemic areas.
Collapse
Affiliation(s)
- Matthew B Laurens
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore 21201, Maryland, USA; .,Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.,Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore 21201, Maryland, USA
| |
Collapse
|
167
|
Guindo A, Sagara I, Ouedraogo B, Sallah K, Assadou MH, Healy S, Duffy P, Doumbo OK, Dicko A, Giorgi R, Gaudart J. "Spatial heterogeneity of environmental risk in randomized prevention trials: consequences and modeling". BMC Med Res Methodol 2019; 19:149. [PMID: 31307393 PMCID: PMC6632226 DOI: 10.1186/s12874-019-0759-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 05/21/2019] [Indexed: 11/30/2022] Open
Abstract
Background In the context of environmentally influenced communicable diseases, proximity to environmental sources results in spatial heterogeneity of risk, which is sometimes difficult to measure in the field. Most prevention trials use randomization to achieve comparability between groups, thus failing to account for heterogeneity. This study aimed to determine under what conditions spatial heterogeneity biases the results of randomized prevention trials, and to compare different approaches to modeling this heterogeneity. Methods Using the example of a malaria prevention trial, simulations were performed to quantify the impact of spatial heterogeneity and to compare different models. Simulated scenarios combined variation in baseline risk, a continuous protective factor (age), a non-related factor (sex), and a binary protective factor (preventive treatment). Simulated spatial heterogeneity scenarios combined variation in breeding site density and effect, location, and population density. The performances of the following five statistical models were assessed: a non-spatial Cox Proportional Hazard (Cox-PH) model and four models accounting for spatial heterogeneity—i.e., a Data-Generating Model, a Generalized Additive Model (GAM), and two Stochastic Partial Differential Equation (SPDE) models, one modeling survival time and the other the number of events. Using a Bayesian approach, we estimated the SPDE models with an Integrated Nested Laplace Approximation algorithm. For each factor (age, sex, treatment), model performances were assessed by quantifying parameter estimation biases, mean square errors, confidence interval coverage rates (CRs), and significance rates. The four models were applied to data from a malaria transmission blocking vaccine candidate. Results The level of baseline risk did not affect our estimates. However, with a high breeding site density and a strong breeding site effect, the Cox-PH and GAM models underestimated the age and treatment effects (but not the sex effect) with a low CR. When population density was low, the Cox-SPDE model slightly overestimated the effect of related factors (age, treatment). The two SPDE models corrected the impact of spatial heterogeneity, thus providing the best estimates. Conclusion Our results show that when spatial heterogeneity is important but not measured, randomization alone cannot achieve comparability between groups. In such cases, prevention trials should model spatial heterogeneity with an adapted method. Trial registration The dataset used for the application example was extracted from Vaccine Trial #NCT02334462 (ClinicalTrials.gov registry). Electronic supplementary material The online version of this article (10.1186/s12874-019-0759-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Abdoulaye Guindo
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, Marseille, France. .,Malaria Research and Training Center - Ogobara K Doumbo, FMOS-FAPH, Mali-NIAID-ICER, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali.
| | - Issaka Sagara
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, Marseille, France.,Malaria Research and Training Center - Ogobara K Doumbo, FMOS-FAPH, Mali-NIAID-ICER, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Boukary Ouedraogo
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, Marseille, France.,Direction des systèmes d'information en santé, Ministère de la santé, Ouagadougou, Burkina Faso
| | - Kankoe Sallah
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, Marseille, France.,AP-HP, Hôpital Bichat, Unité de Recherche Clinique PNVS, Paris, France
| | - Mahamadoun Hamady Assadou
- Malaria Research and Training Center - Ogobara K Doumbo, FMOS-FAPH, Mali-NIAID-ICER, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Sara Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Patrick Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Ogobara K Doumbo
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, Marseille, France.,Malaria Research and Training Center - Ogobara K Doumbo, FMOS-FAPH, Mali-NIAID-ICER, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Alassane Dicko
- Malaria Research and Training Center - Ogobara K Doumbo, FMOS-FAPH, Mali-NIAID-ICER, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Roch Giorgi
- Aix Marseille Univ, APHM, INSERM, IRD, SESSTIM, Hop Timone, BioSTIC, Biostatistic & ICT, Marseille, France
| | - Jean Gaudart
- Aix Marseille Univ, APHM, INSERM, IRD, SESSTIM, Hop Timone, BioSTIC, Biostatistic & ICT, Marseille, France
| |
Collapse
|
168
|
Frimpong A, Kusi KA, Adu-Gyasi D, Amponsah J, Ofori MF, Ndifon W. Phenotypic Evidence of T Cell Exhaustion and Senescence During Symptomatic Plasmodium falciparum Malaria. Front Immunol 2019; 10:1345. [PMID: 31316497 PMCID: PMC6611412 DOI: 10.3389/fimmu.2019.01345] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 05/28/2019] [Indexed: 01/07/2023] Open
Abstract
T cells play significant roles during Plasmodium falciparum infections. Their regulation of the immune response in symptomatic children with malaria has been deemed necessary to prevent immune associated pathology. In this study, we phenotypically characterized the expression of T cell inhibitory(PD-1, CTLA-4) and senescent markers (CD28(-), CD57) from children with symptomatic malaria, asymptomatic malaria and healthy controls using flow cytometry. We observed increased expression of T cell exhaustion and senescence markers in the symptomatic children compared to the asymptomatic and healthy controls. T cell senescence markers were more highly expressed on CD8 T cells than on CD4 T cells. Asymptomatically infected children had comparable levels of these markers with healthy controls except for CD8+ PD-1+ T cells which were significantly elevated in the asymptomatic children. Also, using multivariate regression analysis, CTLA-4 was the only marker that could predict parasitaemia level. The results suggest that the upregulation of immune exhaustion and senescence markers during symptomatic malaria may affect the effector function of T cells leading to inefficient clearance of parasites, hence the inability to develop sterile immunity to malaria.
Collapse
Affiliation(s)
- Augustina Frimpong
- Department of Biochemistry, Cell, and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.,Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana.,African Institute for Mathematical Sciences, Cape Coast, Ghana
| | - Kwadwo Asamoah Kusi
- Department of Biochemistry, Cell, and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.,Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | | | - Jones Amponsah
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Michael Fokuo Ofori
- Department of Biochemistry, Cell, and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.,Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Wilfred Ndifon
- African Institute for Mathematical Sciences, Cape Coast, Ghana.,African Institute for Mathematical Sciences, University of Stellenbosch, Cape Town, South Africa
| |
Collapse
|
169
|
Goh YS, McGuire D, Rénia L. Vaccination With Sporozoites: Models and Correlates of Protection. Front Immunol 2019; 10:1227. [PMID: 31231377 PMCID: PMC6560154 DOI: 10.3389/fimmu.2019.01227] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Despite continuous efforts, the century-old goal of eradicating malaria still remains. Multiple control interventions need to be in place simultaneously to achieve this goal. In addition to effective control measures, drug therapies and insecticides, vaccines are critical to reduce mortality and morbidity. Hence, there are numerous studies investigating various malaria vaccine candidates. Most of the malaria vaccine candidates are subunit vaccines. However, they have shown limited efficacy in Phase II and III studies. To date, only whole parasite formulations have been shown to induce sterile immunity in human. In this article, we review and discuss the recent developments in vaccination with sporozoites and the mechanisms of protection involved.
Collapse
Affiliation(s)
- Yun Shan Goh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore
| | - Daniel McGuire
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
170
|
Barry A, Behet MC, Nébié I, Lanke K, Grignard L, Ouedraogo A, Soulama I, Drakeley C, Sauerwein R, Bolscher JM, Dechering KJ, Bousema T, Tiono AB, Gonçalves BP. Functional antibodies against Plasmodium falciparum sporozoites are associated with a longer time to qPCR-detected infection among schoolchildren in Burkina Faso. Wellcome Open Res 2019; 3:159. [PMID: 30828645 PMCID: PMC6381444 DOI: 10.12688/wellcomeopenres.14932.2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2019] [Indexed: 11/23/2022] Open
Abstract
Background: Individuals living in malaria-endemic regions develop immunity against severe malaria, but it is unclear whether immunity against pre-erythrocytic stages that blocks initiation of blood-stage infection after parasite inoculation develops following continuous natural exposure. Methods: We cleared schoolchildren living in an area (health district of Saponé, Burkina Faso) with highly endemic seasonal malaria of possible sub-patent infections and examined them weekly for incident infections by nested PCR. Plasma samples collected at enrolment were used to quantify antibodies to the pre-eryhrocytic-stage antigens circumsporozoite protein (CSP) and Liver stage antigen 1 (LSA-1).
In vitro sporozoite gliding inhibition and hepatocyte invasion inhibition by naturally acquired antibodies were assessed using
Plasmodium falciparum NF54 sporozoites. Associations between antibody responses, functional pre-erythrocytic immunity phenotypes and time to infection detected by
18S quantitative PCR were studied. Results: A total of 51 children were monitored. Anti-CSP antibody titres showed a positive association with sporozoite gliding motility inhibition (P<0.0001, Spearman’s ρ=0.76).
In vitro hepatocyte invasion was inhibited by naturally acquired antibodies (median inhibition, 19.4% [IQR 15.2-40.9%]), and there were positive correlations between invasion inhibition and gliding inhibition (P=0.005, Spearman’s ρ=0.67) and between invasion inhibition and CSP-specific antibodies (P=0.002, Spearman’s ρ=0.76). Survival analysis indicated longer time to infection in individuals displaying higher-than-median sporozoite gliding inhibition activity (P=0.01), although this association became non-significant after adjustment for blood-stage immunity (P = 0.06). Conclusions: In summary, functional antibodies against the pre-erythrocytic stages of malaria infection are acquired in children who are repeatedly exposed to
Plasmodium parasites. This immune response does not prevent them from becoming infected during a malaria transmission season, but might delay the appearance of blood stage parasitaemia. Our approach could not fully separate the effects of pre-erythrocytic-specific and blood-stage-specific antibody-mediated immune responses
in vivo; epidemiological studies powered and designed to address this important question should become a research priority.
Collapse
Affiliation(s)
- Aissata Barry
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso.,Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marije C Behet
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Issa Nébié
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Kjerstin Lanke
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lynn Grignard
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Alphonse Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Issiaka Soulama
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Chris Drakeley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Robert Sauerwein
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Teun Bousema
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Alfred B Tiono
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Bronner P Gonçalves
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
171
|
Barry A, Behet MC, Nébié I, Lanke K, Grignard L, Ouedraogo A, Soulama I, Drakeley C, Sauerwein R, Bolscher JM, Dechering KJ, Bousema T, Tiono AB, Gonçalves BP. Functional antibodies against Plasmodium falciparum sporozoites are associated with a longer time to qPCR-detected infection among schoolchildren in Burkina Faso. Wellcome Open Res 2019; 3:159. [PMID: 30828645 DOI: 10.12688/wellcomeopenres.14932.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2018] [Indexed: 01/29/2023] Open
Abstract
Background: Individuals living in malaria-endemic regions develop immunity against severe malaria, but it is unclear whether immunity against pre-erythrocytic stages that blocks initiation of blood-stage infection after parasite inoculation develops following continuous natural exposure. Methods: We cleared schoolchildren living in an area (health district of Saponé, Burkina Faso) with highly endemic seasonal malaria of possible sub-patent infections and examined them weekly for incident infections by nested PCR. Plasma samples collected at enrolment were used to quantify antibodies to the pre-eryhrocytic-stage antigens circumsporozoite protein (CSP) and Liver stage antigen 1 (LSA-1). In vitro sporozoite gliding inhibition and hepatocyte invasion inhibition by naturally acquired antibodies were assessed using Plasmodium falciparum NF54 sporozoites. Associations between antibody responses, functional pre-erythrocytic immunity phenotypes and time to infection detected by 18S quantitative PCR were studied. Results: A total of 51 children were monitored. Anti-CSP antibody titres showed a positive association with sporozoite gliding motility inhibition (P<0.0001, Spearman's ρ=0.76). In vitro hepatocyte invasion was inhibited by naturally acquired antibodies (median inhibition, 19.4% [IQR 15.2-40.9%]), and there were positive correlations between invasion inhibition and gliding inhibition (P=0.005, Spearman's ρ=0.67) and between invasion inhibition and CSP-specific antibodies (P=0.002, Spearman's ρ=0.76). Survival analysis indicated longer time to infection in individuals displaying higher-than-median sporozoite gliding inhibition activity (P=0.01), although this association became non-significant after adjustment for blood-stage immunity (P = 0.06). Conclusions: In summary, functional antibodies against the pre-erythrocytic stages of malaria infection are acquired in children who are repeatedly exposed to Plasmodium parasites. This immune response does not prevent them from becoming infected during a malaria transmission season, but might delay the appearance of blood stage parasitaemia. Our approach could not fully separate the effects of pre-erythrocytic-specific and blood-stage-specific antibody-mediated immune responses in vivo; epidemiological studies powered and designed to address this important question should become a research priority.
Collapse
Affiliation(s)
- Aissata Barry
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso.,Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marije C Behet
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Issa Nébié
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Kjerstin Lanke
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lynn Grignard
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Alphonse Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Issiaka Soulama
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Chris Drakeley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Robert Sauerwein
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Teun Bousema
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Alfred B Tiono
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Bronner P Gonçalves
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
172
|
Schindler T, Jongo S, Studer F, Mpina M, Mwangoka G, Mswata S, Ramadhani K, Sax J, Church LWP, Richie TL, Tanner M, Hoffman SL, Abdulla S, Daubenberger C. Two cases of long-lasting, sub-microscopic Plasmodium malariae infections in adults from coastal Tanzania. Malar J 2019; 18:149. [PMID: 31036014 PMCID: PMC6489319 DOI: 10.1186/s12936-019-2787-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/22/2019] [Indexed: 01/29/2023] Open
Abstract
Background Malaria is endemic in Tanzania with majority of clinical cases caused by Plasmodium falciparum. Additionally, Plasmodium malariae and Plasmodium ovale spp. are also present and clinical manifestations caused by these infections are not well described. Clinical episodes caused by P. malariae infections are often characterized by a relatively mild illness with a low number of parasites, which can persist for long periods. In this report, two cases of P. malariae infections that were identified during a clinical trial evaluating the P. falciparum malaria vaccine candidate, PfSPZ Vaccine are described. The two participants were followed up and monitored for clinical and laboratory parameters to assess vaccine safety providing the opportunity to study clinical manifestations of P. malariae over 4 months. Case presentation Two young, healthy Tanzanian men infected with low density asexual blood stage P. malariae diagnosed by quantitative polymerase chain reaction (qPCR) are described. Retrospective analysis of collected and stored blood samples revealed that the two volunteers had constant asexual blood stage parasitaemia for more than 4 months. During the 132 days of infection, the volunteers’ vital signs, body temperature and serum biochemistry all remained within normal ranges. Haematological abnormalities, which were transiently outside normal ranges, were regarded as not clinically significant. During this time period, four consecutive evaluations of blood samples by thick blood smear microscopy conducted by an experienced microscopist were all negative, indicating the presence of low-density sub-microscopic infections. Conclusions The two cases of P. malariae infections presented here confirm the ability of this Plasmodium species to persist at low density in the human host for extended time periods without causing clinical symptoms. The presented data also demonstrate that clinical study sites in malaria endemic regions need to have a strong malaria diagnostic infrastructure, including the ability of capturing sub-microscopic parasitaemia and differentiation of Plasmodium species. Trial registration ClinicalTrials.gov: NCT02613520, https://clinicaltrials.gov/ct2/show/NCT02613520, Registered: November 24th 2015, Enrolment of the first participant to the trial: December 15th 2015, Trial was registered before the first participant was enrolled
Collapse
Affiliation(s)
- Tobias Schindler
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| | - Said Jongo
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, United Republic of Tanzania.
| | - Fabian Studer
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Maximilian Mpina
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland.,Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, United Republic of Tanzania
| | - Grace Mwangoka
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, United Republic of Tanzania
| | - Sarah Mswata
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, United Republic of Tanzania
| | - Kamaka Ramadhani
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, United Republic of Tanzania
| | - Julian Sax
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | | | | | - Marcel Tanner
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | | | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, United Republic of Tanzania
| | - Claudia Daubenberger
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| |
Collapse
|
173
|
Diallo H, Katile A, Kwan JL, Sissoko MS, Healy SA, Doumbo OK, Duffy PE, Zaidi I. Longitudinal analysis of gamma delta T cell subsets during malaria infections in Malian adults. Malar J 2019; 18:69. [PMID: 30866943 PMCID: PMC6416881 DOI: 10.1186/s12936-019-2702-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/01/2019] [Indexed: 02/07/2023] Open
Abstract
Background Immunity that limits malarial disease is acquired over time, but adults living in endemic areas continue to become infected and can require treatment for clinical illness. Gamma delta (γδ) T cells, particularly the Vδ2+ subset, have been associated with development of clinical malaria in children. In this study, the dynamics of total γδ T cells, Vδ2+ and Vδ2− T cells were measured during a malaria transmission season in Malian adults. Methods This study explored γδ T cell dynamics and Plasmodium falciparum infection outcomes over the course of the malaria transmission season in Malian adults enrolled in the placebo arm of a double-blind randomized vaccine trial. All volunteers were treated with anti-malarial drugs prior to the start of the transmission season and blood smears were assessed for P. falciparum infection every 2 weeks from July 2014 to January 2015. The study participants were stratified as either asymptomatic infections or clinical malaria cases. Vδ2+ and Vδ2− γδ T cell frequencies and activation (as measured by CD38 expression) were measured in all study participants at baseline and then every 2 months using a whole blood flow cytometry assay. Results Forty of the forty-three subjects became infected with P. falciparum and, of those, 21 individuals were diagnosed with clinical malaria at least once during the season. The γδ T cell percentage and activation increased over the duration of the transmission season. Both the Vδ2+ and Vδ2− γδ T cells were activated by P. falciparum infection. Conclusion γδ T cells increased during a malaria transmission season and this expansion was noted in both the Vδ2+ and Vδ2− γδ T cells. However, neither expansion or activation of either γδ T cell subsets discriminated study participants that had asymptomatic infections from those that had clinical malaria cases.
Collapse
Affiliation(s)
- Hama Diallo
- Malaria Research Training Center, International Center of Excellence in Research, Faculty of Medicine and Pharmacy of the University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Abdoulaye Katile
- Malaria Research Training Center, International Center of Excellence in Research, Faculty of Medicine and Pharmacy of the University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Jennifer L Kwan
- Laboratory for Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Mahamadou S Sissoko
- Malaria Research Training Center, International Center of Excellence in Research, Faculty of Medicine and Pharmacy of the University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Sara A Healy
- Laboratory for Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Ogobara K Doumbo
- Malaria Research Training Center, International Center of Excellence in Research, Faculty of Medicine and Pharmacy of the University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Patrick E Duffy
- Laboratory for Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| | - Irfan Zaidi
- Laboratory for Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
174
|
Cooper MM, Loiseau C, McCarthy JS, Doolan DL. Human challenge models: tools to accelerate the development of malaria vaccines. Expert Rev Vaccines 2019; 18:241-251. [PMID: 30732492 DOI: 10.1080/14760584.2019.1580577] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Malaria challenge models, where healthy human volunteers are intentionally infected with Plasmodium species parasites under controlled conditions, can be undertaken in several well-defined ways. These challenge models enable evaluation of the kinetics of parasite growth and clearance, host-pathogen interactions and the host immune response. They can facilitate discovery of candidate diagnostic biomarkers and novel vaccine targets. As translational tools they can facilitate testing of candidate vaccines and drugs and evaluation of diagnostic tests. AREAS COVERED Until recently, malaria human challenge models have been limited to only a few Plasmodium falciparum strains and used exclusively in malaria-naïve volunteers in non-endemic regions. Several recent advances include the use of alternate P. falciparum strains and other species of Plasmodia, as well as strains attenuated by chemical, radiation or genetic modification, and the conduct of studies in pre-exposed individuals. Herein, we discuss how this diversification is enabling more thorough vaccine efficacy testing and informing rational vaccine development. EXPERT OPINION The ability to comprehensively evaluate vaccine efficacy in controlled settings will continue to accelerate the translation of candidate malaria vaccines to the clinic, and inform the development and optimisation of potential vaccines that would be effective against multiple strains in geographically and demographically diverse settings.
Collapse
Affiliation(s)
- Martha M Cooper
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - Claire Loiseau
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - James S McCarthy
- b Infectious Diseases Programme , QIMR Berghofer Medical Research Institute , Brisbane , Australia
| | - Denise L Doolan
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| |
Collapse
|
175
|
Abstract
The development of highly effective and durable vaccines against the human malaria parasites Plasmodium falciparum and P. vivax remains a key priority. Decades of endeavor have taught that achieving this goal will be challenging; however, recent innovation in malaria vaccine research and a diverse pipeline of novel vaccine candidates for clinical assessment provides optimism. With first-generation pre-erythrocytic vaccines aiming for licensure in the coming years, it is important to reflect on how next-generation approaches can improve on their success. Here we review the latest vaccine approaches that seek to prevent malaria infection, disease, and transmission and highlight some of the major underlying immunological and molecular mechanisms of protection. The synthesis of rational antigen selection, immunogen design, and immunization strategies to induce quantitatively and qualitatively improved immune effector mechanisms offers promise for achieving sustained high-level protection.
Collapse
|
176
|
Ssemaganda A, Giddam AK, Zaman M, Skwarczynski M, Toth I, Stanisic DI, Good MF. Induction of Plasmodium-Specific Immune Responses Using Liposome-Based Vaccines. Front Immunol 2019; 10:135. [PMID: 30774635 PMCID: PMC6367261 DOI: 10.3389/fimmu.2019.00135] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/16/2019] [Indexed: 12/30/2022] Open
Abstract
In the development of vaccines, the ability to initiate both innate and subsequent adaptive immune responses need to be considered. Live attenuated vaccines achieve this naturally, while inactivated and sub-unit vaccines generally require additional help provided through delivery systems and/or adjuvants. Liposomes present an attractive adjuvant/delivery system for antigens. Here, we review the key aspects of immunity against Plasmodium parasites, liposome design considerations and their current application in the development of a malaria vaccine.
Collapse
Affiliation(s)
| | | | - Mehfuz Zaman
- Institute for Glycomics, Griffith University, Southport, QLD, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | | | - Michael F. Good
- Institute for Glycomics, Griffith University, Southport, QLD, Australia
| |
Collapse
|
177
|
Wilson KL, Flanagan KL, Prakash MD, Plebanski M. Malaria vaccines in the eradication era: current status and future perspectives. Expert Rev Vaccines 2019; 18:133-151. [PMID: 30601095 DOI: 10.1080/14760584.2019.1561289] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The challenge to eradicate malaria is an enormous task that will not be achieved by current control measures, thus an efficacious and long-lasting malaria vaccine is required. The licensing of RTS, S/AS01 is a step forward in providing some protection, but a malaria vaccine that protects across multiple transmission seasons is still needed. To achieve this, inducing beneficial immune responses while minimising deleterious non-targeted effects will be essential. AREAS COVERED This article discusses the current challenges and advances in malaria vaccine development and reviews recent human clinical trials for each stage of infection. Pubmed and ScienceDirect were searched, focusing on cell mediated immunity and how T cell subsets might be targeted in future vaccines using novel adjuvants and emerging vaccine technologies. EXPERT COMMENTARY Despite decades of research there is no highly effective licensed malaria vaccine. However, there is cause for optimism as new adjuvants and vaccine systems emerge, and our understanding of correlates of protection increases, especially regarding cellular immunity. The new field of heterologous (non-specific) effects of vaccines also highlights the broader consequences of immunization. Importantly, the WHO led Malaria Vaccine Technology Roadmap illustrates that there is a political will among the global health community to make it happen.
Collapse
Affiliation(s)
- K L Wilson
- a Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences , Monash University , Melbourne , Australia.,b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| | - K L Flanagan
- a Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences , Monash University , Melbourne , Australia.,b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia.,c School of Medicine, Faculty of Health Sciences , University of Tasmania , Launceston , Australia
| | - M D Prakash
- b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| | - M Plebanski
- b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| |
Collapse
|
178
|
Beeson JG, Kurtovic L, Dobaño C, Opi DH, Chan JA, Feng G, Good MF, Reiling L, Boyle MJ. Challenges and strategies for developing efficacious and long-lasting malaria vaccines. Sci Transl Med 2019; 11:11/474/eaau1458. [DOI: 10.1126/scitranslmed.aau1458] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/05/2018] [Accepted: 11/02/2018] [Indexed: 12/24/2022]
Abstract
Although there has been major recent progress in malaria vaccine development, substantial challenges remain for achieving highly efficacious and durable vaccines against Plasmodium falciparum and Plasmodium vivax malaria. Greater knowledge of mechanisms and key targets of immunity are needed to accomplish this goal, together with new strategies for generating potent, long-lasting, functional immunity against multiple antigens. Implementation considerations in endemic areas will ultimately affect vaccine effectiveness, so innovations to simplify and enhance delivery are also needed. Whereas challenges remain, recent exciting progress and emerging knowledge promise hope for the future of malaria vaccines.
Collapse
|
179
|
Healy SA, Fried M, Richie T, Bok K, Little M, August A, Riley L, Swamy GK, Wylie BJ, Menendez C, Muehlenbachs A, Doumbo O, Greenwood B, Billingsley PF, Hoffman SL, Duffy PE. Malaria vaccine trials in pregnant women: An imperative without precedent. Vaccine 2019; 37:763-770. [PMID: 30621913 DOI: 10.1016/j.vaccine.2018.12.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 12/11/2018] [Accepted: 12/13/2018] [Indexed: 11/28/2022]
Abstract
Pregnant women are highly susceptible to Plasmodium falciparum malaria, leading to substantial maternal, perinatal, and infant mortality. While malaria vaccine development has made significant progress in recent years, no trials of malaria vaccines have ever been conducted in pregnant women. In December 2016, an expert meeting was convened at NIAID, NIH, in Rockville, Maryland to deliberate on the rationale and design of malaria vaccine trials in pregnant women. The discussions highlighted the progress made over recent years in the field of maternal immunization for other infectious diseases, and the evolving regulatory and ethical environment, all of which support a new emphasis on testing malaria vaccines that offer direct benefits to pregnant women. Initial safety and immunogenicity studies of malaria vaccines will be conducted in non-pregnant adult volunteers. Subsequently, efficacy trials involving pregnant women will likely be conducted in malaria-endemic and often resource-poor environments where sufficiently high malaria incidence will allow vaccine activity to be measured. Such trials will need to meet all international standards to ensure the safety of mother and offspring, under oversight of appropriate ethical and regulatory bodies. The convened experts drafted a clinical development plan to test a malaria vaccine product during pregnancy, using as a case study PfSPZ Vaccine being developed by Sanaria Inc. that is currently in phase 2 testing. Following the expert recommendations, a pregnancy registry has been initiated in Ouelessebougou, Mali, to provide baseline information on maternal and fetal outcomes as a context for evaluating PfSPZ Vaccine safety in the future, and new regimens are being assessed that will be suitable for evaluation in pregnant women.
Collapse
Affiliation(s)
- Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Karin Bok
- National Vaccine Program Office (NVPO), Office of Assistant Secretary for Health, Washington, D.C., USA
| | - Maggie Little
- Kennedy Institute of Ethics, Georgetown University, Washington, D.C., USA
| | - Allison August
- Clinical Development, Moderna Therapeutics, Cambridge, MA, USA
| | - Laura Riley
- Obstetrics, Massachusetts General Hospital, Boston, MA, USA
| | - Geeta K Swamy
- Department of Obstetrics & Gynecology, Duke University, Durham, NC, USA
| | - Blair J Wylie
- Maternal-Fetal Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Clara Menendez
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic-Universitat de Barcelona, Spain
| | | | - Ogobara Doumbo
- Malaria Research and Training Center, Université de Bamako, Mali, Bamako, Mali
| | | | | | | | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
180
|
O'Brochta DA, Alford R, Harrell R, Aluvihare C, Eappen AG, Li T, Chakravarty S, Sim BKL, Hoffman SL, Billingsley PF. Is Saglin a mosquito salivary gland receptor for Plasmodium falciparum? Malar J 2019; 18:2. [PMID: 30602380 PMCID: PMC6317240 DOI: 10.1186/s12936-018-2634-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/20/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Saglin, a 100 kDa protein composed of two 50 kDa homodimers, is present in the salivary glands of Anopheles gambiae and has been considered an essential receptor for sporozoites (SPZ) of Plasmodium berghei and Plasmodium falciparum (Pf), allowing SPZ to recognize, bind to, and infect mosquito salivary glands. Spatial and temporal patterns of Saglin expression reported here, however, suggest that this model does not fully describe the Saglin-SPZ interaction. RESULTS Saglin protein was detected by indirect immunofluorescence microscopy only in the medial and proximal-lateral lobes, but not in the distal-lateral lobes, of the salivary glands of An. gambiae; the pattern of expression was independent of mosquito age or physiological state. These results were confirmed by steady-state Saglin transcript and protein expression using qRT-PCR and Western-blot analysis, respectively. Saglin was localized to the basal surface of the cells of the medial lobes and was undetectable elsewhere (intracellularly, on the lateral or apical membranes, the cells' secretory vacuoles, or in the salivary duct). In the cells of the proximal lateral lobes of the salivary glands, Saglin was distinctly intracellular and was not localized to any of the cell surfaces. Transgenic Anopheles stephensi were produced that expressed An. gambiae Saglin in the distal lateral lobes of the salivary gland. Additional Saglin expression did not enhance infection by PfSPZ compared to non-transgenic siblings fed on the same gametocyte-containing blood meal. CONCLUSIONS The absence of Saglin in the distal lateral lobes of the salivary glands, a primary destination for SPZ, suggests Saglin is not an essential receptor for Plasmodium SPZ. The lack of any correlation between increased Saglin expression in the distal lateral lobes of the salivary glands of transgenic An. stephensi and PfSPZ infection is also consistent with Saglin not being an essential salivary gland receptor for Plasmodium SPZ.
Collapse
Affiliation(s)
- David A O'Brochta
- Department of Entomology and The Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, 9600 Gudelsky Drive, Rockville, MD, 20850, USA.,Foundation for the National Institutes of Health, 11400 Rockville Pike, Suite 600, North Bethesda, MD, 20852, USA
| | - Robert Alford
- Department of Entomology and The Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Robert Harrell
- University of Maryland Insect Transformation Facility, The Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Channa Aluvihare
- University of Maryland Insect Transformation Facility, The Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD, 20850, USA
| | - Abraham G Eappen
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Tao Li
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Sumana Chakravarty
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - B Kim Lee Sim
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Stephen L Hoffman
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Peter F Billingsley
- Sanaria Inc., 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA.
| |
Collapse
|
181
|
Ricotta E, Kwan J. Artemisinin-Resistant Malaria as a Global Catastrophic Biological Threat. Curr Top Microbiol Immunol 2019; 424:33-57. [PMID: 31218504 DOI: 10.1007/82_2019_163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The global spread of artemisinin resistance brings with it the threat of incurable malaria. Already, this disease threatens over 219 million lives per year and causes 5-6% losses in GDP in endemic areas, even with current advances in prevention and treatment. This chapter discusses the currently tenuous position we are in globally, and the impact that could be seen if artemisinin treatment is lost, whether due to the unchecked spread of K13 mutations or poor global investment in treatment and prevention advances. Artemisinin is the backbone of current ACT treatment programs and severe malarial treatment; without it, the success of future malaria eradication programs will be in jeopardy.
Collapse
Affiliation(s)
- Emily Ricotta
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Quarters 15B-1, 8 West Dr, Bethesda, MD, 20892, USA.
- Kelly Government Solutions, Bethesda, USA.
| | - Jennifer Kwan
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Quarters 15B-1, 8 West Dr, Bethesda, MD, 20892, USA
| |
Collapse
|
182
|
Deroost K, Langhorne J. Gamma/Delta T Cells and Their Role in Protection Against Malaria. Front Immunol 2018; 9:2973. [PMID: 30619330 PMCID: PMC6306408 DOI: 10.3389/fimmu.2018.02973] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/03/2018] [Indexed: 12/28/2022] Open
Abstract
Whether and how γδT cells play a protective role in immunity against Plasmodium infection remain open questions. γδT cells expand in patients and mice infected with Plasmodium spp, and cytokine production and cytotoxic responses against blood-stage parasites are observed in vitro. Their expansion is associated with protective immunity induced by irradiated sporozoite immunization, and depletion of γδT cells in some mouse models of malaria excacerbates blood-stage infections. It is now clear that these cells can have many different functions, and data are emerging suggesting that in addition to having direct parasitocidal effects, they can regulate other immune cells during Plasmodium infections. Here we review some of the historic and more recent data on γδT cells, and in light of the new information on their potential protective roles we suggest that it is a good time to re-evaluate their activation requirements, specificity and function during malaria.
Collapse
|
183
|
Tan J, Piccoli L, Lanzavecchia A. The Antibody Response to Plasmodium falciparum: Cues for Vaccine Design and the Discovery of Receptor-Based Antibodies. Annu Rev Immunol 2018; 37:225-246. [PMID: 30566366 DOI: 10.1146/annurev-immunol-042617-053301] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Plasmodium falciparum remains a serious public health problem and a continuous challenge for the immune system due to the complexity and diversity of the pathogen. Recent advances from several laboratories in the characterization of the antibody response to the parasite have led to the identification of critical targets for protection and revealed a new mechanism of diversification based on the insertion of host receptors into immunoglobulin genes, leading to the production of receptor-based antibodies. These advances have opened new possibilities for vaccine design and passive antibody therapies to provide sterilizing immunity and control blood-stage parasites.
Collapse
Affiliation(s)
- Joshua Tan
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland; .,Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, United Kingdom.,Current affiliation: National Institute of Allergy and Infectious Diseases, Rockville, Maryland 20852, USA
| | - Luca Piccoli
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland;
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland; .,VIR Biotechnology, San Francisco, California 94158, USA
| |
Collapse
|
184
|
Tiono AB, Nébié I, Anagnostou N, Coulibaly AS, Bowyer G, Lam E, Bougouma EC, Ouedraogo A, Yaro JBB, Barry A, Roberts R, Rampling T, Bliss C, Hodgson S, Lawrie A, Ouedraogo A, Imoukhuede EB, Ewer KJ, Viebig NK, Diarra A, Leroy O, Bejon P, Hill AVS, Sirima SB. First field efficacy trial of the ChAd63 MVA ME-TRAP vectored malaria vaccine candidate in 5-17 months old infants and children. PLoS One 2018; 13:e0208328. [PMID: 30540808 PMCID: PMC6291132 DOI: 10.1371/journal.pone.0208328] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/13/2018] [Indexed: 01/21/2023] Open
Abstract
Background Heterologous prime boost immunization with chimpanzee adenovirus 63 (ChAd63) and Modified Vaccinia Virus Ankara (MVA) vectored vaccines is a strategy previously shown to provide substantial protective efficacy against P. falciparum infection in United Kingdom adult Phase IIa sporozoite challenge studies (approximately 20–25% sterile protection with similar numbers showing clear delay in time to patency), and greater point efficacy in a trial in Kenyan adults. Methodology We conducted the first Phase IIb clinical trial assessing the safety, immunogenicity and efficacy of ChAd63 MVA ME-TRAP in 700 healthy malaria exposed children aged 5–17 months in a highly endemic malaria transmission area of Burkina Faso. Results ChAd63 MVA ME-TRAP was shown to be safe and immunogenic but induced only moderate T cell responses (median 326 SFU/106 PBMC (95% CI 290–387)) many fold lower than in previous trials. No significant efficacy was observed against clinical malaria during the follow up period, with efficacy against the primary endpoint estimate by proportional analysis being 13.8% (95%CI -42.4 to 47.9) at sixth month post MVA ME-TRAP and 3.1% (95%CI -15.0 to 18.3; p = 0.72) by Cox regression. Conclusions This study has confirmed ChAd63 MVA ME-TRAP is a safe and immunogenic vaccine regimen in children and infants with prior exposure to malaria. But no significant protective efficacy was observed in this very highly malaria-endemic setting. Trial registration ClinicalTrials.gov NCT01635647. Pactr.org PACTR201208000404131.
Collapse
Affiliation(s)
- Alfred B. Tiono
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Issa Nébié
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Nicholas Anagnostou
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Aboubacar S. Coulibaly
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Georgina Bowyer
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Erika Lam
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Edith C. Bougouma
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Alphonse Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Jean Baptist B. Yaro
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Aïssata Barry
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Rachel Roberts
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Tommy Rampling
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Carly Bliss
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Susanne Hodgson
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Alison Lawrie
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Amidou Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | | | - Katie J. Ewer
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicola K. Viebig
- European Vaccine Initiative, Universitäts Klinikum Heidelberg, Heidelberg, Germany
| | - Amidou Diarra
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Odile Leroy
- European Vaccine Initiative, Universitäts Klinikum Heidelberg, Heidelberg, Germany
| | - Philip Bejon
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Adrian V. S. Hill
- The Jenner Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Sodiomon B. Sirima
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
- * E-mail:
| |
Collapse
|
185
|
Itsara LS, Zhou Y, Do J, Grieser AM, Vaughan AM, Ghosh AK. The Development of Whole Sporozoite Vaccines for Plasmodium falciparum Malaria. Front Immunol 2018; 9:2748. [PMID: 30619241 PMCID: PMC6297750 DOI: 10.3389/fimmu.2018.02748] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/08/2018] [Indexed: 12/20/2022] Open
Abstract
Each year malaria kills hundreds of thousands of people and infects hundreds of millions of people despite current control measures. An effective malaria vaccine will likely be necessary to aid in malaria eradication. Vaccination using whole sporozoites provides an increased repertoire of immunogens compared to subunit vaccines across at least two life cycle stages of the parasite, the extracellular sporozoite, and intracellular liver stage. Three potential whole sporozoite vaccine approaches are under development and include genetically attenuated parasites, radiation attenuated sporozoites, and wild-type sporozoites administered in combination with chemoprophylaxis. Pre-clinical and clinical studies have demonstrated whole sporozoite vaccine immunogenicity, including humoral and cellular immunity and a range of vaccine efficacy that depends on the pre-exposure of vaccinated individuals. While whole sporozoite vaccines can provide protection against malaria in some cases, more recent studies in malaria-endemic regions demonstrate the need for improvements. Moreover, challenges remain in manufacturing large quantities of sporozoites for vaccine commercialization. A promising solution to the whole sporozoite manufacturing challenge is in vitro culturing methodology, which has been described for several Plasmodium species, including the major disease-causing human malaria parasite, Plasmodium falciparum. Here, we review whole sporozoite vaccine immunogenicity and in vitro culturing platforms for sporozoite production.
Collapse
Affiliation(s)
| | | | - Julie Do
- MalarVx, Inc., Seattle, WA, United States
| | | | - Ashley M Vaughan
- Seattle Children's Research Institute, Seattle, WA, United States
| | | |
Collapse
|
186
|
Kapulu MC, Njuguna P, Hamaluba MM. Controlled Human Malaria Infection in Semi-Immune Kenyan Adults (CHMI-SIKA): a study protocol to investigate in vivo Plasmodium falciparum malaria parasite growth in the context of pre-existing immunity. Wellcome Open Res 2018; 3:155. [PMID: 31803847 PMCID: PMC6871356 DOI: 10.12688/wellcomeopenres.14909.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2018] [Indexed: 10/20/2023] Open
Abstract
Malaria remains a major public health burden despite approval for implementation of a partially effective pre-erythrocytic malaria vaccine. There is an urgent need to accelerate development of a more effective multi-stage vaccine. Adults in malaria endemic areas may have substantial immunity provided by responses to the blood stages of malaria parasites, but field trials conducted on several blood-stage vaccines have not shown high levels of efficacy. We will use controlled human malaria infection (CHMI) studies with malaria-exposed volunteers to identify correlations between immune responses and parasite growth rates in vivo. Immune responses more strongly associated with control of parasite growth should be prioritized to accelerate malaria vaccine development. We aim to recruit up to 200 healthy adult volunteers from areas of differing malaria transmission in Kenya, and after confirming their health status through clinical examination and routine haematology and biochemistry, we will comprehensively characterize immunity to malaria using >100 blood-stage antigens. We will administer 3,200 aseptic, purified, cryopreserved Plasmodium falciparum sporozoites (PfSPZ Challenge) by direct venous inoculation. Serial quantitative polymerase chain reaction to measure parasite growth rate in vivo will be undertaken. Clinical and laboratory monitoring will be undertaken to ensure volunteer safety. In addition, we will also explore the perceptions and experiences of volunteers and other stakeholders in participating in a malaria volunteer infection study. Serum, plasma, peripheral blood mononuclear cells and extracted DNA will be stored to allow a comprehensive assessment of adaptive and innate host immunity. We will use CHMI in semi-immune adult volunteers to relate parasite growth outcomes with antibody responses and other markers of host immunity. Registration: ClinicalTrials.gov identifier NCT02739763.
Collapse
Affiliation(s)
- Melissa C. Kapulu
- KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, UK
| | | | | | - CHMI-SIKA Study Team
- KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University Oxford, Oxford, UK
| |
Collapse
|
187
|
Frimpong A, Kusi KA, Ofori MF, Ndifon W. Novel Strategies for Malaria Vaccine Design. Front Immunol 2018; 9:2769. [PMID: 30555463 PMCID: PMC6281765 DOI: 10.3389/fimmu.2018.02769] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022] Open
Abstract
The quest for a licensed effective vaccine against malaria remains a global priority. Even though classical vaccine design strategies have been successful for some viral and bacterial pathogens, little success has been achieved for Plasmodium falciparum, which causes the deadliest form of malaria due to its diversity and ability to evade host immune responses. Nevertheless, recent advances in vaccinology through high throughput discovery of immune correlates of protection, lymphocyte repertoire sequencing and structural design of immunogens, provide a comprehensive approach to identifying and designing a highly efficacious vaccine for malaria. In this review, we discuss novel vaccine approaches that can be employed in malaria vaccine design.
Collapse
Affiliation(s)
- Augustina Frimpong
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.,African Institute for Mathematical Sciences, Cape Coast, Ghana
| | - Kwadwo Asamoah Kusi
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Michael Fokuo Ofori
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Wilfred Ndifon
- African Institute for Mathematical Sciences, Cape Coast, Ghana.,African Institute for Mathematical Sciences, University of Stellenbosch, Cape Town, South Africa
| |
Collapse
|
188
|
Pre-clinical evaluation of a P. berghei-based whole-sporozoite malaria vaccine candidate. NPJ Vaccines 2018; 3:54. [PMID: 30510775 PMCID: PMC6258718 DOI: 10.1038/s41541-018-0091-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/17/2018] [Indexed: 11/11/2022] Open
Abstract
Whole-sporozoite vaccination/immunization induces high levels of protective immunity in both rodent models of malaria and in humans. Recently, we generated a transgenic line of the rodent malaria parasite P. berghei (Pb) that expresses the P. falciparum (Pf) circumsporozoite protein (PfCS), and showed that this parasite line (PbVac) was capable of (1) infecting and developing in human hepatocytes but not in human erythrocytes, and (2) inducing neutralizing antibodies against the human Pf parasite. Here, we analyzed PbVac in detail and developed tools necessary for its use in clinical studies. A microbiological contaminant-free Master Cell Bank of PbVac parasites was generated through a process of cyclic propagation and clonal expansion in mice and mosquitoes and was genetically characterized. A highly sensitive qRT-PCR-based method was established that enables PbVac parasite detection and quantification at low parasite densities in vivo. This method was employed in a biodistribution study in a rabbit model, revealing that the parasite is only present at the site of administration and in the liver up to 48 h post infection and is no longer detectable at any site 10 days after administration. An extensive toxicology investigation carried out in rabbits further showed the absence of PbVac-related toxicity. In vivo drug sensitivity assays employing rodent models of infection showed that both the liver and the blood stage forms of PbVac were completely eliminated by Malarone® treatment. Collectively, our pre-clinical safety assessment demonstrates that PbVac possesses all characteristics necessary to advance into clinical evaluation. PbVac is a transgenic malaria parasite expressing circumsporozoite antigen from the human parasite Plasmodium falciparum. PbVac elicits neutralizing P. falciparum antibodies and can infect human hepatocytes but not erythrocytes, suggesting that humans would be non-permissive. Miguel Prudêncio and colleagues at the Institute of Molecular Medicine in Lisbon perform a detailed in vivo analysis and toxicology of PbVac. Extensive biodistribution analysis using a highly sensitive qPCR in non-permissive rabbit hosts shows PbVac are present at the initial bite site early on with later appearance in the liver, but by day 10 is undetectable. Importantly no PbVac could be detected in the blood at any time-point. PbVac was well tolerated with no apparent pathological signatures. In permissive mouse hosts PbVac could be effectively eliminated from both the blood and liver and could thereby act as a potential clinical ‘safety net’ in the event of an erythrocytic stage or persistence in the liver.
Collapse
|
189
|
Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat Immunol 2018; 19:1199-1211. [PMID: 30333613 DOI: 10.1038/s41590-018-0228-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/31/2018] [Indexed: 02/08/2023]
Abstract
Development of a malaria vaccine remains a critical priority to decrease clinical disease and mortality and facilitate eradication. Accordingly, RTS,S, a protein-subunit vaccine, has completed phase III clinical trials and confers ~30% protection against clinical infection over 4 years. Whole-attenuated-sporozoite and viral-subunit vaccines induce between 20% and 100% protection against controlled human malaria infection, but there is limited published evidence to date for durable, high-level efficacy (>50%) against natural exposure. Importantly, fundamental scientific advances related to the potency, durability, breadth and location of immune responses will be required for improving vaccine efficacy with these and other vaccine approaches. In this Review, we focus on the current understanding of immunological mechanisms of protection from animal models and human vaccine studies, and on how these data should inform the development of next-generation vaccines. Furthermore, we introduce the concept of using passive immunization with monoclonal antibodies as a new approach to prevent and eliminate malaria.
Collapse
Affiliation(s)
- Ian A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
190
|
Xia L, Wu J, Pattaradilokrat S, Tumas K, He X, Peng YC, Huang R, Myers TG, Long CA, Wang R, Su XZ. Detection of host pathways universally inhibited after Plasmodium yoelii infection for immune intervention. Sci Rep 2018; 8:15280. [PMID: 30327482 PMCID: PMC6191451 DOI: 10.1038/s41598-018-33599-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/26/2018] [Indexed: 12/16/2022] Open
Abstract
Malaria is a disease with diverse symptoms depending on host immune status and pathogenicity of Plasmodium parasites. The continuous parasite growth within a host suggests mechanisms of immune evasion by the parasite and/or immune inhibition in response to infection. To identify pathways commonly inhibited after malaria infection, we infected C57BL/6 mice with four Plasmodium yoelii strains causing different disease phenotypes and 24 progeny of a genetic cross. mRNAs from mouse spleens day 1 and/or day 4 post infection (p.i.) were hybridized to a mouse microarray to identify activated or inhibited pathways, upstream regulators, and host genes playing an important role in malaria infection. Strong interferon responses were observed after infection with the N67 strain, whereas initial inhibition and later activation of hematopoietic pathways were found after infection with 17XNL parasite, showing unique responses to individual parasite strains. Inhibitions of pathways such as Th1 activation, dendritic cell (DC) maturation, and NFAT immune regulation were observed in mice infected with all the parasite strains day 4 p.i., suggesting universally inhibited immune pathways. As a proof of principle, treatment of N67-infected mice with antibodies against T cell receptors OX40 or CD28 to activate the inhibited pathways enhanced host survival. Controlled activation of these pathways may provide important strategies for better disease management and for developing an effective vaccine.
Collapse
Affiliation(s)
- Lu Xia
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA.,State Key Laboratory of Medical Genetics, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, The People's Republic of China
| | - Jian Wu
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Sittiporn Pattaradilokrat
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA.,Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Keyla Tumas
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Xiao He
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Yu-Chih Peng
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Timothy G Myers
- Genomic Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Carole A Long
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Rongfu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Xin-Zhuan Su
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA.
| |
Collapse
|
191
|
Stanisic DI, Fink J, Mayer J, Coghill S, Gore L, Liu XQ, El-Deeb I, Rodriguez IB, Powell J, Willemsen NM, De SL, Ho MF, Hoffman SL, Gerrard J, Good MF. Vaccination with chemically attenuated Plasmodium falciparum asexual blood-stage parasites induces parasite-specific cellular immune responses in malaria-naïve volunteers: a pilot study. BMC Med 2018; 16:184. [PMID: 30293531 PMCID: PMC6174572 DOI: 10.1186/s12916-018-1173-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/11/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The continuing morbidity and mortality associated with infection with malaria parasites highlights the urgent need for a vaccine. The efficacy of sub-unit vaccines tested in clinical trials in malaria-endemic areas has thus far been disappointing, sparking renewed interest in the whole parasite vaccine approach. We previously showed that a chemically attenuated whole parasite asexual blood-stage vaccine induced CD4+ T cell-dependent protection against challenge with homologous and heterologous parasites in rodent models of malaria. METHODS In this current study, we evaluated the immunogenicity and safety of chemically attenuated asexual blood-stage Plasmodium falciparum (Pf) parasites in eight malaria-naïve human volunteers. Study participants received a single dose of 3 × 107 Pf pRBC that had been treated in vitro with the cyclopropylpyrolloindole analogue, tafuramycin-A. RESULTS We demonstrate that Pf asexual blood-stage parasites that are completely attenuated are immunogenic, safe and well tolerated in malaria-naïve volunteers. Following vaccination with a single dose, species and strain transcending Plasmodium-specific T cell responses were induced in recipients. This included induction of Plasmodium-specific lymphoproliferative responses, T cells secreting the parasiticidal cytokines, IFN-γ and TNF, and CD3+CD45RO+ memory T cells. Pf-specific IgG was not detected. CONCLUSIONS This is the first clinical study evaluating a whole parasite blood-stage malaria vaccine. Following administration of a single dose of completely attenuated Pf asexual blood-stage parasites, Plasmodium-specific T cell responses were induced while Pf-specific antibodies were not detected. These results support further evaluation of this chemically attenuated vaccine in humans. TRIAL REGISTRATION Trial registration: ACTRN12614000228684 . Registered 4 March 2014.
Collapse
Affiliation(s)
- Danielle I Stanisic
- Institute for Glycomics, Griffith University, Parklands Drive, Southport, Queensland, Australia.
| | - James Fink
- Gold Coast University Hospital, 1 Hospital Blvd, Southport, Queensland, Australia
| | - Johanna Mayer
- Gold Coast University Hospital, 1 Hospital Blvd, Southport, Queensland, Australia
| | - Sarah Coghill
- Gold Coast University Hospital, 1 Hospital Blvd, Southport, Queensland, Australia
| | - Letitia Gore
- Gold Coast University Hospital, 1 Hospital Blvd, Southport, Queensland, Australia
| | - Xue Q Liu
- Institute for Glycomics, Griffith University, Parklands Drive, Southport, Queensland, Australia
| | - Ibrahim El-Deeb
- Institute for Glycomics, Griffith University, Parklands Drive, Southport, Queensland, Australia
| | - Ingrid B Rodriguez
- Institute for Glycomics, Griffith University, Parklands Drive, Southport, Queensland, Australia
| | - Jessica Powell
- Institute for Glycomics, Griffith University, Parklands Drive, Southport, Queensland, Australia
| | - Nicole M Willemsen
- Institute for Glycomics, Griffith University, Parklands Drive, Southport, Queensland, Australia
| | - Sai Lata De
- Institute for Glycomics, Griffith University, Parklands Drive, Southport, Queensland, Australia
| | - Mei-Fong Ho
- Institute for Glycomics, Griffith University, Parklands Drive, Southport, Queensland, Australia
| | | | - John Gerrard
- Gold Coast University Hospital, 1 Hospital Blvd, Southport, Queensland, Australia
| | - Michael F Good
- Institute for Glycomics, Griffith University, Parklands Drive, Southport, Queensland, Australia.
| |
Collapse
|
192
|
McCall MBB, Kremsner PG, Mordmüller B. Correlating efficacy and immunogenicity in malaria vaccine trials. Semin Immunol 2018; 39:52-64. [PMID: 30219621 DOI: 10.1016/j.smim.2018.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 12/19/2022]
Abstract
The availability of an effective and appropriately implemented malaria vaccine would form a crucial cornerstone of public health efforts to fight this disease. Despite many decades of research, however, no malaria vaccine has yet shown satisfactory protective efficacy or been rolled-out. Validated immunological substitute endpoints have the potential to accelerate clinical vaccine development by reducing the required complexity, size, duration and cost of clinical trials. Besides facilitating clinical development of existing vaccine candidates, understanding immunological mechanisms of protection may drive the development of fundamentally new vaccination approaches. In this review we focus on correlates of protection in malaria vaccine development: Does immunogenicity predict malaria vaccine efficacy and why is this question particularly difficult? Have immunological correlates accelerated malaria vaccine development in the past and will they facilitate it in the future? Does Controlled Human Malaria Infection represent a valid model for identifying such immunological correlates, or a correlate of protection against naturally-acquired malaria in itself?
Collapse
Affiliation(s)
- Matthew B B McCall
- Institut für Tropenmedizin, Universität Tübingen and Deutsches Zentrum für Infektionsforschung, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.
| | - Peter G Kremsner
- Institut für Tropenmedizin, Universität Tübingen and Deutsches Zentrum für Infektionsforschung, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Benjamin Mordmüller
- Institut für Tropenmedizin, Universität Tübingen and Deutsches Zentrum für Infektionsforschung, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
193
|
Abstract
In malaria, the immune responses leading to protective immunity versus immunopathology are unclear. Mamedov et al. (2018) identify a subset of clonally expanded γδ T cells in late-stage infection that produce M-CSF and may interact with myeloid cells to control recrudescent infection.
Collapse
|
194
|
Coelho CH. Malaria vaccines as prevention strategies: for more studies on community perception. Malawi Med J 2018; 30:140. [PMID: 30627345 PMCID: PMC6307060 DOI: 10.4314/mmj.v30i3.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Camila H Coelho
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, USA
| |
Collapse
|
195
|
Mendes AM, Machado M, Gonçalves-Rosa N, Reuling IJ, Foquet L, Marques C, Salman AM, Yang ASP, Moser KA, Dwivedi A, Hermsen CC, Jiménez-Díaz B, Viera S, Santos JM, Albuquerque I, Bhatia SN, Bial J, Angulo-Barturen I, Silva JC, Leroux-Roels G, Janse CJ, Khan SM, Mota MM, Sauerwein RW, Prudêncio M. A Plasmodium berghei sporozoite-based vaccination platform against human malaria. NPJ Vaccines 2018; 3:33. [PMID: 30155278 PMCID: PMC6109154 DOI: 10.1038/s41541-018-0068-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/21/2018] [Accepted: 05/31/2018] [Indexed: 12/15/2022] Open
Abstract
There is a pressing need for safe and highly effective Plasmodium falciparum (Pf) malaria vaccines. The circumsporozoite protein (CS), expressed on sporozoites and during early hepatic stages, is a leading target vaccine candidate, but clinical efficacy has been modest so far. Conversely, whole-sporozoite (WSp) vaccines have consistently shown high levels of sterilizing immunity and constitute a promising approach to effective immunization against malaria. Here, we describe a novel WSp malaria vaccine that employs transgenic sporozoites of rodent P. berghei (Pb) parasites as cross-species immunizing agents and as platforms for expression and delivery of PfCS (PbVac). We show that both wild-type Pb and PbVac sporozoites unabatedly infect and develop in human hepatocytes while unable to establish an infection in human red blood cells. In a rabbit model, similarly susceptible to Pb hepatic but not blood infection, we show that PbVac elicits cross-species cellular immune responses, as well as PfCS-specific antibodies that efficiently inhibit Pf sporozoite liver invasion in human hepatocytes and in mice with humanized livers. Thus, PbVac is safe and induces functional immune responses in preclinical studies, warranting clinical testing and development. A genetically engineered parasite, related to malaria-causing Plasmodium falciparum, excels as a vaccine in preclinical tests. A team led by Miguel Prudêncio, of the University of Lisbon, Portugal, developed a genetically altered vaccine candidate based on Plasmodium berghei, which is pathogenic to rodents but, in humans, fails to progress from a harmless, transient liver infection to causing full, blood-borne malaria. The candidate expresses a human form of ‘circumsporozoite protein,’ a known antigen, and is designed to provoke a more comprehensive immune response as it presents a whole pathogen to the host. In preclinical tests, the candidate generated antibodies able to neutralize infection in human hepatocytes and also provoked a cellular immune response in rabbits. The team’s candidate proved safe and efficacious, warranting further trials and clinical testing.
Collapse
Affiliation(s)
- António M Mendes
- 1Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Marta Machado
- 1Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Nataniel Gonçalves-Rosa
- 1Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Isaie J Reuling
- 2Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB Nijmegen, The Netherlands
| | - Lander Foquet
- 3Center for Vaccinology, Ghent University and Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.,Departments of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Cláudia Marques
- 1Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Ahmed M Salman
- 5Leiden Malaria Research Group, Parasitology, Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands.,6The Jenner Institute, Nuffield Department of Medicine, University of Oxford, ORCRB, Roosevelt Drive, Oxford, OX3 7DQ UK
| | - Annie S P Yang
- 2Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB Nijmegen, The Netherlands
| | - Kara A Moser
- 7Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Ankit Dwivedi
- 7Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Cornelus C Hermsen
- 2Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB Nijmegen, The Netherlands
| | - Belén Jiménez-Díaz
- 8Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa, 2, 28760 Tres Cantos, Madrid Spain
| | - Sara Viera
- 8Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa, 2, 28760 Tres Cantos, Madrid Spain
| | - Jorge M Santos
- 1Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal.,12Present Address: Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, 02115 Boston, MA USA
| | - Inês Albuquerque
- 1Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Sangeeta N Bhatia
- 9Health Sciences and Technology/Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - John Bial
- 10Yecuris Corporation, PO Box 4645, Tualatin, OR 97062 USA
| | - Iñigo Angulo-Barturen
- 8Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa, 2, 28760 Tres Cantos, Madrid Spain
| | - Joana C Silva
- 7Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201 USA.,11Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Geert Leroux-Roels
- 3Center for Vaccinology, Ghent University and Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Chris J Janse
- 5Leiden Malaria Research Group, Parasitology, Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Shahid M Khan
- 5Leiden Malaria Research Group, Parasitology, Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria M Mota
- 1Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Robert W Sauerwein
- 2Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB Nijmegen, The Netherlands
| | - Miguel Prudêncio
- 1Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
196
|
Othman AS, Franke-Fayard BM, Imai T, van der Gracht ETI, Redeker A, Salman AM, Marin-Mogollon C, Ramesar J, Chevalley-Maurel S, Janse CJ, Arens R, Khan SM. OX40 Stimulation Enhances Protective Immune Responses Induced After Vaccination With Attenuated Malaria Parasites. Front Cell Infect Microbiol 2018; 8:247. [PMID: 30073152 PMCID: PMC6060232 DOI: 10.3389/fcimb.2018.00247] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/28/2018] [Indexed: 01/15/2023] Open
Abstract
Protection against a malaria infection can be achieved by immunization with live-attenuated Plasmodium sporozoites and while the precise mechanisms of protection remain unknown, T cell responses are thought to be critical in the elimination of infected liver cells. In cancer immunotherapies, agonistic antibodies that target T cell surface proteins, such as CD27, OX40 (CD134), and 4-1BB (CD137), have been used to enhance T cell function by increasing co-stimulation. In this study, we have analyzed the effect of agonistic OX40 monoclonal antibody treatment on protective immunity induced in mice immunized with genetically attenuated parasites (GAPs). OX40 stimulation enhanced protective immunity after vaccination as shown by an increase in the number of protected mice and delay to blood-stage infection after challenge with wild-type sporozoites. Consistent with the enhanced protective immunity enforced OX40 stimulation resulted in an increased expansion of antigen-experienced effector (CD11ahiCD44hi) CD8+ and CD4+ T cells in the liver and spleen and also increased IFN-γ and TNF producing CD4+ T cells in the liver and spleen. In addition, GAP immunization plus α-OX40 treatment significantly increased sporozoite-specific IgG responses. Thus, we demonstrate that targeting T cell costimulatory receptors can improve sporozoite-based vaccine efficacy.
Collapse
Affiliation(s)
- Ahmad Syibli Othman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Blandine M Franke-Fayard
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Takashi Imai
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Esmé T I van der Gracht
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Anke Redeker
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Ahmed M Salman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands.,The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Catherin Marin-Mogollon
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Jai Ramesar
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Chris J Janse
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Shahid M Khan
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
197
|
Frimpong A, Kusi KA, Tornyigah B, Ofori MF, Ndifon W. Characterization of T cell activation and regulation in children with asymptomatic Plasmodium falciparum infection. Malar J 2018; 17:263. [PMID: 30005684 PMCID: PMC6045887 DOI: 10.1186/s12936-018-2410-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/07/2018] [Indexed: 02/08/2023] Open
Abstract
Background Asymptomatic Plasmodium infections are characterized by the absence of clinical disease and the ability to restrict parasite replication. Increasing levels of regulatory T cells (Tregs) in Plasmodium falciparum infections have been associated with the risk of developing clinical disease, suggesting that individuals with asymptomatic infections may have reduced Treg frequency. However, the relationship between Tregs, cellular activation and parasite control in asymptomatic malaria remains unclear. Methods In a cross-sectional study, the levels of Tregs and other T cell activation phenotypes were compared using flow cytometry in symptomatic, asymptomatic and uninfected children before and after stimulation with infected red blood cell lysates (iRBCs). In addition, the association between these T cell phenotypes and parasitaemia were investigated. Results In children with asymptomatic infections, levels of Tregs and activated T cells were comparable to those in healthy controls but significantly lower than those in symptomatic children. After iRBC stimulation, levels of Tregs remained lower for asymptomatic versus symptomatic children. In contrast, levels of activated T cells were higher for asymptomatic children. Strikingly, the pre-stimulation levels of two T cell activation phenotypes (CD8+CD69+ and CD8+CD25+CD69+) and the post-stimulation levels of two regulatory phenotypes (CD4+CD25+Foxp3+ and CD8+CD25+Foxp3+) were significantly positively correlated with and explained 68% of the individual variation in parasitaemia. A machine-learning model based on levels of these four phenotypes accurately distinguished between asymptomatic and symptomatic children (sensitivity = 86%, specificity = 94%), suggesting that these phenotypes govern the observed variation in disease status. Conclusion Compared to symptomatic P. falciparum infections, in children asymptomatic infections are characterized by lower levels of Tregs and activated T cells, which are associated with lower parasitaemia. The results indicate that T cell regulatory and activation phenotypes govern both parasitaemia and disease status in paediatric malaria in the studied sub-Saharan African population. Electronic supplementary material The online version of this article (10.1186/s12936-018-2410-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Augustina Frimpong
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, P. O. Box LG 54, Accra, Ghana. .,Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P.O. Box LG 581, Accra, Ghana. .,African Institute for Mathematical Sciences, P.O. Box DL 676, Cape-Coast, Ghana.
| | - Kwadwo Asamoah Kusi
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, P. O. Box LG 54, Accra, Ghana.,Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P.O. Box LG 581, Accra, Ghana
| | - Bernard Tornyigah
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P.O. Box LG 581, Accra, Ghana
| | - Michael Fokuo Ofori
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, P. O. Box LG 54, Accra, Ghana.,Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P.O. Box LG 581, Accra, Ghana
| | - Wilfred Ndifon
- African Institute for Mathematical Sciences, P.O. Box DL 676, Cape-Coast, Ghana. .,African Institute for Mathematical Sciences, University of Stellenbosch, 7 Melrose Rd, Muizenberg, Cape Town, 7945, South Africa.
| |
Collapse
|
198
|
Ratto-Kim S, Yoon IK, Paris RM, Excler JL, Kim JH, O’Connell RJ. The US Military Commitment to Vaccine Development: A Century of Successes and Challenges. Front Immunol 2018; 9:1397. [PMID: 29977239 PMCID: PMC6021486 DOI: 10.3389/fimmu.2018.01397] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/05/2018] [Indexed: 12/04/2022] Open
Abstract
The US military has been a leading proponent of vaccine development since its founding. General George Washington ordered the entire American army to be variolated against smallpox after recognizing the serious threat that it posed to military operations. He did this on the recommendation from Dr. John Morgan, the physician-in-chief of the American army, who wrote a treatise on variolation in 1776. Although cases of smallpox still occurred, they were far fewer than expected, and it is believed that the vaccination program contributed to victory in the War of Independence. Effective military force requires personnel who are healthy and combat ready for worldwide deployment. Given the geography of US military operations, military personnel should also be protected against diseases that are endemic in potential areas of conflict. For this reason, and unknown to many, the US military has strongly supported vaccine research and development. Four categories of communicable infectious diseases threaten military personnel: (1) diseases that spread easily in densely populated areas (respiratory and dysenteric diseases); (2) vector-borne diseases (disease carried by mosquitoes and other insects); (3) sexually transmitted diseases (hepatitis, HIV, and gonorrhea); and (4) diseases associated with biological warfare. For each category, the US military has supported research that has provided the basis for many of the vaccines available today. Although preventive measures and the development of drugs have provided some relief from the burden of malaria, dengue, and HIV, the US military continues to fund research and development of prophylactic vaccines that will contribute to force health protection and global health. In the past few years, newly recognized infections with Zika, severe acute respiratory syndrome, Middle East respiratory syndrome viruses have pushed the US military to fund research and fast track clinical trials to quickly and effectively develop vaccines for emerging diseases. With US military personnel present in every region of the globe, one of the most cost-effective ways to maintain military effectiveness is to develop vaccines against prioritized threats to military members' health.
Collapse
Affiliation(s)
| | - In-Kyu Yoon
- International Vaccine Institute, Seoul, South Korea
| | | | | | | | | |
Collapse
|
199
|
Jongo SA, Shekalaghe SA, Church LWP, Ruben AJ, Schindler T, Zenklusen I, Rutishauser T, Rothen J, Tumbo A, Mkindi C, Mpina M, Mtoro AT, Ishizuka AS, Kassim KR, Milando FA, Qassim M, Juma OA, Mwakasungula S, Simon B, James ER, Abebe Y, Kc N, Chakravarty S, Saverino E, Bakari BM, Billingsley PF, Seder RA, Daubenberger C, Sim BKL, Richie TL, Tanner M, Abdulla S, Hoffman SL. Safety, Immunogenicity, and Protective Efficacy against Controlled Human Malaria Infection of Plasmodium falciparum Sporozoite Vaccine in Tanzanian Adults. Am J Trop Med Hyg 2018; 99:338-349. [PMID: 29943719 PMCID: PMC6090339 DOI: 10.4269/ajtmh.17-1014] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
We are using controlled human malaria infection (CHMI) by direct venous inoculation (DVI) of cryopreserved, infectious Plasmodium falciparum (Pf) sporozoites (SPZ) (PfSPZ Challenge) to try to reduce time and costs of developing PfSPZ Vaccine to prevent malaria in Africa. Immunization with five doses at 0, 4, 8, 12, and 20 weeks of 2.7 × 105 PfSPZ of PfSPZ Vaccine gave 65% vaccine efficacy (VE) at 24 weeks against mosquito bite CHMI in U.S. adults and 52% (time to event) or 29% (proportional) VE over 24 weeks against naturally transmitted Pf in Malian adults. We assessed the identical regimen in Tanzanians for VE against PfSPZ Challenge. Twenty- to thirty-year-old men were randomized to receive five doses normal saline or PfSPZ Vaccine in a double-blind trial. Vaccine efficacy was assessed 3 and 24 weeks later. Adverse events were similar in vaccinees and controls. Antibody responses to Pf circumsporozoite protein were significantly lower than in malaria-naïve Americans, but significantly higher than in Malians. All 18 controls developed Pf parasitemia after CHMI. Four of 20 (20%) vaccinees remained uninfected after 3 week CHMI (P = 0.015 by time to event, P = 0.543 by proportional analysis) and all four (100%) were uninfected after repeat 24 week CHMI (P = 0.005 by proportional, P = 0.004 by time to event analysis). Plasmodium falciparum SPZ Vaccine was safe, well tolerated, and induced durable VE in four subjects. Controlled human malaria infection by DVI of PfSPZ Challenge appeared more stringent over 24 weeks than mosquito bite CHMI in United States or natural exposure in Malian adults, thereby providing a rigorous test of VE in Africa.
Collapse
Affiliation(s)
- Said A Jongo
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Seif A Shekalaghe
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | | | | | - Tobias Schindler
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - Isabelle Zenklusen
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - Tobias Rutishauser
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - Julian Rothen
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - Anneth Tumbo
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Catherine Mkindi
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Maximillian Mpina
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Ali T Mtoro
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Andrew S Ishizuka
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - Florence A Milando
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Munira Qassim
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Omar A Juma
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Solomon Mwakasungula
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Beatus Simon
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | | | | | | | | | | | - Bakari M Bakari
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | | | - Robert A Seder
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Claudia Daubenberger
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - B Kim Lee Sim
- Protein Potential LLC, Rockville, Maryland.,Sanaria Inc., Rockville, Maryland
| | | | - Marcel Tanner
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute (Swiss TPH), Basel, Switzerland
| | - Salim Abdulla
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | | |
Collapse
|
200
|
Rothen J, Murie C, Carnes J, Anupama A, Abdulla S, Chemba M, Mpina M, Tanner M, Lee Sim BK, Hoffman SL, Gottardo R, Daubenberger C, Stuart K. Whole blood transcriptome changes following controlled human malaria infection in malaria pre-exposed volunteers correlate with parasite prepatent period. PLoS One 2018; 13:e0199392. [PMID: 29920562 PMCID: PMC6007927 DOI: 10.1371/journal.pone.0199392] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/29/2018] [Indexed: 12/15/2022] Open
Abstract
Malaria continues to be one of mankind’s most devastating diseases despite the many and varied efforts to combat it. Indispensable for malaria elimination and eventual eradication is the development of effective vaccines. Controlled human malaria infection (CHMI) is an invaluable tool for vaccine efficacy assessment and investigation of early immunological and molecular responses against Plasmodium falciparum infection. Here, we investigated gene expression changes following CHMI using RNA-Seq. Peripheral blood samples were collected in Bagamoyo, Tanzania, from ten adults who were injected intradermally (ID) with 2.5x104 aseptic, purified, cryopreserved P. falciparum sporozoites (Sanaria® PfSPZ Challenge). A total of 2,758 genes were identified as differentially expressed following CHMI. Transcriptional changes were most pronounced on day 5 after inoculation, during the clinically silent liver phase. A secondary analysis, grouping the volunteers according to their prepatent period duration, identified 265 genes whose expression levels were linked to time of blood stage parasitemia detection. Gene modules associated with these 265 genes were linked to regulation of transcription, cell cycle, phosphatidylinositol signaling and erythrocyte development. Our study showed that in malaria pre-exposed volunteers, parasite prepatent period in each individual is linked to magnitude and timing of early gene expression changes after ID CHMI.
Collapse
Affiliation(s)
- Julian Rothen
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Carl Murie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jason Carnes
- Center for Infectious Disease Research, Seattle, Washington, United States of America
| | - Atashi Anupama
- Center for Infectious Disease Research, Seattle, Washington, United States of America
| | - Salim Abdulla
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Mwajuma Chemba
- Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Maxmillian Mpina
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland.,Bagamoyo Research and Training Centre, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Marcel Tanner
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - B Kim Lee Sim
- Sanaria Inc., Rockville, Maryland, United States of America
| | | | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Claudia Daubenberger
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Ken Stuart
- Center for Infectious Disease Research, Seattle, Washington, United States of America
| |
Collapse
|