1
|
de Franca MNF, Rodrigues LS, Barreto AS, da Cruz GS, Aragão-Santos JC, da Silva AM, de Jesus AR, Palatnik-de-Sousa CB, de Almeida RP, Corrêa CB. CD4 + Th1 and Th17 responses and multifunctional CD8 T lymphocytes associated with cure or disease worsening in human visceral leishmaniasis. Front Immunol 2024; 15:1277557. [PMID: 38410517 PMCID: PMC10895669 DOI: 10.3389/fimmu.2024.1277557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/02/2024] [Indexed: 02/28/2024] Open
Abstract
Introduction In VL, a proinflammatory phenotype is typically associated with enhanced phagocytosis and a Th1 mediated immune response resulting in infection control. In contrast, an anti-inflammatory phenotype, associated with a predominant regulatory response, typically enables intracellular multiplication of Leishmania parasites and disease progression. Methods To investigate the impact of chemotherapy on Th2 and Th17 immune responses in patients with visceral leishmaniasis (VL), we assessed all combinations of intracellular expression of IFN-γ, IL-10, IL-4 and IL-17 in the CD4+ and CD8+ T cell populations of peripheral blood mononuclear cell (PBMC) samples from patients, after antigenic stimulation with Leishmania lysate, throughout treatment and follow-up. As increases in spleen and liver sizes and decreases in hematocrit, hemogloblin, erythrocytes, monocytes, leukocytes and platelets levels are strongly related to the disease, we studied the correlations between the frequencies of T cells producing the afore mentioned cytokines, individually and in combination, and these variables, as markers of disease or cure. Results We found that the frequency of IFN-γ-producingCD4+ T cells increased until the end of chemotherapy with Glucantime® or AmBisome ®, while IL-10, IL-4 and IL-17-producing CD4+ T cells peaked on day 7 following the start of treatment. Although the frequency of CD4+IL-17+ cells decreased during treatment an increase was observed after clinical cure. The frequency of CD4+ T cells producing only IFN-γ or IL-17 correlated with blood monocytes levels. Frequencies of double-producers of IFN-γ and IL-10 or IL-4 correlated positively with eosinophils and platelets levels. Together, this suggest that IFN-γ drives the immune response towards Th1 at cure. In contrast, and associated with disease or Th2 response, the frequency of CD4+ IL-10+ cells correlated positively with spleen sizes and negatively with circulating monocyte levels, while the frequency of CD4+ producing both IL-4 and IL-10 correlated negatively with platelets levels. The frequency of CD8+ single-producers of IFN-γ increased from day 21 to 90 while that of single-producers of IL-10 peaked on day 7, of IL-4 on day 30 and of IL-17, on day 180. IFN-γ expression in CD8+ single- and double-producers of cytokines was indicative of an immune response associated with cure. In contrast, frequencies of CD8+ double-producers of IL-4 and IL-10, IL-4 and IL-17 and IL-10 and IL-17 and producers of three and four cytokines, were associated with disease and were low after the cure. Frequencies of CD8+ T cells producing IFN-γ alone or with IL-17 were positively correlated with platelets levels. In contrast, as markers of disease: 1) frequencies of single producers of IL-10 correlated negatively with leukocytes levels, 2) frequencies of double producers of IL-4 and IL-10 correlated negatively with platelet, leukocyte, lymphocyte and circulating monocyte levels, 3) frequencies of triple-producers of IFN-γ, IL-4 and IL-10 correlated negatively with platelet, leukocyte and neutrophil levels and 4) frequencies of producers of IFN-γ, IL-4, IL-10 and IL-17 simultaneously correlated positively with spleen size, and negatively with leukocyte and neutrophil levels. Discussion Our results confirmed that the clinical improvement of VL patients correlates with the decrease of an IL-4 and IL-10 CD4+Th2 response, the recovery of CD4+ Th1 and Th17 responses and the frequency of CD8+ single-producers of IFN-γ and double producers of IFN-γ and IL-17.
Collapse
Affiliation(s)
- Mariana Nobre Farias de Franca
- Laboratory of Biology and Immunology of Cancer and Leishmania, Department of Morphology, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Lorranny Santana Rodrigues
- Laboratory of Biology and Immunology of Cancer and Leishmania, Department of Morphology, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Aline Silva Barreto
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Division of Immunology and Molecular Biology Laboratory, University Hospital/Empresa Brasileira de Serviços Hospitalares (EBSERBH), Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Geydson Silveira da Cruz
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Division of Immunology and Molecular Biology Laboratory, University Hospital/Empresa Brasileira de Serviços Hospitalares (EBSERBH), Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - José Carlos Aragão-Santos
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Department of Physical Education, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Angela Maria da Silva
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Department of Medicine, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Amélia Ribeiro de Jesus
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Division of Immunology and Molecular Biology Laboratory, University Hospital/Empresa Brasileira de Serviços Hospitalares (EBSERBH), Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Department of Medicine, Federal University of Sergipe, Immunology Investigative Institute (III), INCT, CNPq, Aracaju, Sergipe, Brazil
| | - Clarisa B. Palatnik-de-Sousa
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Immunology Investigative Institute (III), Insititutos nacionais de Ciência e Tecnologia (INCT), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Rio de Janeiro, Brazil
- Graduate Program in Vegetal Biotechnology and Bioprocesses, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Roque Pacheco de Almeida
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Division of Immunology and Molecular Biology Laboratory, University Hospital/Empresa Brasileira de Serviços Hospitalares (EBSERBH), Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Department of Medicine, Federal University of Sergipe, Immunology Investigative Institute (III), INCT, CNPq, Aracaju, Sergipe, Brazil
| | - Cristiane Bani Corrêa
- Laboratory of Biology and Immunology of Cancer and Leishmania, Department of Morphology, Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Graduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| |
Collapse
|
2
|
Natama HM, Moncunill G, Vidal M, Rouamba T, Aguilar R, Santano R, Rovira-Vallbona E, Jiménez A, Somé MA, Sorgho H, Valéa I, Coulibaly-Traoré M, Coppel RL, Cavanagh D, Chitnis CE, Beeson JG, Angov E, Dutta S, Gamain B, Izquierdo L, Mens PF, Schallig HDFH, Tinto H, Rosanas-Urgell A, Dobaño C. Associations between prenatal malaria exposure, maternal antibodies at birth, and malaria susceptibility during the first year of life in Burkina Faso. Infect Immun 2023; 91:e0026823. [PMID: 37754682 PMCID: PMC10580994 DOI: 10.1128/iai.00268-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/12/2023] [Indexed: 09/28/2023] Open
Abstract
In this study, we investigated how different categories of prenatal malaria exposure (PME) influence levels of maternal antibodies in cord blood samples and the subsequent risk of malaria in early childhood in a birth cohort study (N = 661) nested within the COSMIC clinical trial (NCT01941264) in Burkina Faso. Plasmodium falciparum infections during pregnancy and infants' clinical malaria episodes detected during the first year of life were recorded. The levels of maternal IgG and IgG1-4 to 15 P. falciparum antigens were measured in cord blood by quantitative suspension array technology. Results showed a significant variation in the magnitude of maternal antibody levels in cord blood, depending on the PME category, with past placental malaria (PM) more frequently associated with significant increases of IgG and/or subclass levels across three groups of antigens defined as pre-erythrocytic, erythrocytic, and markers of PM, as compared to those from the cord of non-exposed control infants. High levels of antibodies to certain erythrocytic antigens (i.e., IgG to EBA140 and EBA175, IgG1 to EBA175 and MSP142, and IgG3 to EBA140 and MSP5) were independent predictors of protection from clinical malaria during the first year of life. By contrast, high levels of IgG, IgG1, and IgG2 to the VAR2CSA DBL1-2 and IgG4 to DBL3-4 were significantly associated with an increased risk of clinical malaria. These findings indicate that PME categories have different effects on the levels of maternal-derived antibodies to malaria antigens in children at birth, and this might drive heterogeneity to clinical malaria susceptibility in early childhood.
Collapse
Affiliation(s)
- Hamtandi Magloire Natama
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Gemma Moncunill
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Marta Vidal
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
| | - Toussaint Rouamba
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Ruth Aguilar
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
| | - Rebeca Santano
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Eduard Rovira-Vallbona
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
| | - Alfons Jiménez
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
- CIBER de Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain
| | - M. Athanase Somé
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Hermann Sorgho
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Innocent Valéa
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Maminata Coulibaly-Traoré
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Ross L. Coppel
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - David Cavanagh
- Centre for Immunity, Infection & Evolution, Institute of Immunology & Infection Research, Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Chetan E. Chitnis
- Malaria Parasite Biology and Vaccines Unit, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris, Paris, France
| | | | - Evelina Angov
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, USA
| | - Sheetij Dutta
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, USA
| | | | - Luis Izquierdo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Petra F. Mens
- Academic Medical Centre at the University of Amsterdam, Amsterdam, the Netherlands
| | | | - Halidou Tinto
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Anna Rosanas-Urgell
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Carlota Dobaño
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| |
Collapse
|
3
|
Genito CJ, Brooks K, Smith A, Ryan E, Soto K, Li Y, Warter L, Dutta S. Protective antibody threshold of RTS,S/AS01 malaria vaccine correlates antigen and adjuvant dose in mouse model. NPJ Vaccines 2023; 8:114. [PMID: 37563255 PMCID: PMC10415390 DOI: 10.1038/s41541-023-00714-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
Mouse models are useful for the early down-selection of malaria vaccine candidates. The Walter Reed Army Institute of Research has optimized a transgenic Plasmodium berghei sporozoite challenge model to compare the efficacy of Plasmodium falciparum circumsporozoite protein (CSP) vaccines. GSK's RTS,S vaccine formulated in the adjuvant AS01 can protect malaria-naïve individuals against malaria. We report that the RTS,S/AS01 vaccine induces high level sterile protection in our mouse model. Down titration of the antigen at a constant AS01 dose revealed a potent antigen dose-sparing effect and the superiority of RTS,S/AS01 over a soluble CSP antigen. RTS,S-mediated protective immunity was associated with a threshold of major repeat antibody titer. Combined titration of the antigen and adjuvant showed that reducing the adjuvant could improve antibody boosting post-3rd vaccination and reduce the threshold antibody concentration required for protection. Mouse models can provide a pathway for preclinical assessment of strategies to improve CSP vaccines against malaria.
Collapse
Affiliation(s)
- Christopher J Genito
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Katherine Brooks
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Alexis Smith
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Emma Ryan
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Kim Soto
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Yuanzhang Li
- Center for Enabling Capabilities, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | | | - Sheetij Dutta
- Structural Vaccinology Laboratory, Biologics Research and Development Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA.
| |
Collapse
|
4
|
Ren H, Jia W, Xie Y, Yu M, Chen Y. Adjuvant physiochemistry and advanced nanotechnology for vaccine development. Chem Soc Rev 2023; 52:5172-5254. [PMID: 37462107 DOI: 10.1039/d2cs00848c] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Vaccines comprising innovative adjuvants are rapidly reaching advanced translational stages, such as the authorized nanotechnology adjuvants in mRNA vaccines against COVID-19 worldwide, offering new strategies to effectively combat diseases threatening human health. Adjuvants are vital ingredients in vaccines, which can augment the degree, extensiveness, and longevity of antigen specific immune response. The advances in the modulation of physicochemical properties of nanoplatforms elevate the capability of adjuvants in initiating the innate immune system and adaptive immunity, offering immense potential for developing vaccines against hard-to-target infectious diseases and cancer. In this review, we provide an essential introduction of the basic principles of prophylactic and therapeutic vaccination, key roles of adjuvants in augmenting and shaping immunity to achieve desired outcomes and effectiveness, and the physiochemical properties and action mechanisms of clinically approved adjuvants for humans. We particularly focus on the preclinical and clinical progress of highly immunogenic emerging nanotechnology adjuvants formulated in vaccines for cancer treatment or infectious disease prevention. We deliberate on how the immune system can sense and respond to the physicochemical cues (e.g., chirality, deformability, solubility, topology, and chemical structures) of nanotechnology adjuvants incorporated in the vaccines. Finally, we propose possible strategies to accelerate the clinical implementation of nanotechnology adjuvanted vaccines, such as in-depth elucidation of nano-immuno interactions, antigen identification and optimization by the deployment of high-dimensional multiomics analysis approaches, encouraging close collaborations among scientists from different scientific disciplines and aggressive exploration of novel nanotechnologies.
Collapse
Affiliation(s)
- Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
5
|
Pendyala G, Calvo‐Calle JM, Moreno A, Kane RS. A multivalent Plasmodium falciparum circumsporozoite protein-based nanoparticle malaria vaccine elicits a robust and durable antibody response against the junctional epitope and the major repeats. Bioeng Transl Med 2023; 8:e10514. [PMID: 37476056 PMCID: PMC10354751 DOI: 10.1002/btm2.10514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Plasmodium falciparum (Pf) malaria continues to cause considerable morbidity and mortality worldwide. The circumsporozoite protein (CSP) is a particularly attractive candidate for designing vaccines that target sporozoites-the first vertebrate stage in a malaria infection. Current PfCSP-based vaccines, however, do not include epitopes that have recently been shown to be the target of potent neutralizing antibodies. We report the design of a SpyCatcher-mi3-nanoparticle-based vaccine presenting multiple copies of a chimeric PfCSP (cPfCSP) antigen that incorporates these important "T1/junctional" epitopes as well as a reduced number of (NANP)n repeats. cPfCSP-SpyCatcher-mi3 was immunogenic in mice eliciting high and durable IgG antibody levels as well as a balanced antibody response against the T1/junctional region and the (NANP)n repeats. Notably, the antibody concentration elicited by immunization was significantly greater than the reported protective threshold defined in a murine challenge model. Refocusing the immune response toward functionally relevant subdominant epitopes to induce a more balanced and durable immune response may enable the design of a more effective second generation PfCSP-based vaccine.
Collapse
Affiliation(s)
- Geetanjali Pendyala
- School of Chemical & Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgia30332USA
| | - J. Mauricio Calvo‐Calle
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts01655USA
| | - Alberto Moreno
- Emory Vaccine Center, Emory National Primate Research CenterEmory UniversityAtlantaGeorgia30329USA
- Division of Infectious Diseases, Department of MedicineEmory UniversityAtlantaGeorgia30303USA
| | - Ravi S. Kane
- School of Chemical & Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgia30332USA
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGeorgia30332USA
| |
Collapse
|
6
|
Brai A, Poggialini F, Pasqualini C, Trivisani CI, Vagaggini C, Dreassi E. Progress towards Adjuvant Development: Focus on Antiviral Therapy. Int J Mol Sci 2023; 24:9225. [PMID: 37298177 PMCID: PMC10253057 DOI: 10.3390/ijms24119225] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
In recent decades, vaccines have been extraordinary resources to prevent pathogen diffusion and cancer. Even if they can be formed by a single antigen, the addition of one or more adjuvants represents the key to enhance the response of the immune signal to the antigen, thus accelerating and increasing the duration and the potency of the protective effect. Their use is of particular importance for vulnerable populations, such as the elderly or immunocompromised people. Despite their importance, only in the last forty years has the search for novel adjuvants increased, with the discovery of novel classes of immune potentiators and immunomodulators. Due to the complexity of the cascades involved in immune signal activation, their mechanism of action remains poorly understood, even if significant discovery has been recently made thanks to recombinant technology and metabolomics. This review focuses on the classes of adjuvants under research, recent mechanism of action studies, as well as nanodelivery systems and novel classes of adjuvants that can be chemically manipulated to create novel small molecule adjuvants.
Collapse
Affiliation(s)
- Annalaura Brai
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Federica Poggialini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Pasqualini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Immacolata Trivisani
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Chiara Vagaggini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Elena Dreassi
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| |
Collapse
|
7
|
Moita D, Maia TG, Duarte M, Andrade CM, Albuquerque IS, Dwivedi A, Silva JC, González-Céron L, Janse CJ, Mendes AM, Prudêncio M. A genetically modified Plasmodium berghei parasite as a surrogate for whole-sporozoite vaccination against P. vivax malaria. NPJ Vaccines 2022; 7:163. [PMID: 36526627 PMCID: PMC9755804 DOI: 10.1038/s41541-022-00585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
Two malaria parasite species, Plasmodium falciparum (Pf) and P. vivax (Pv) are responsible for most of the disease burden caused by malaria. Vaccine development against this disease has focused mainly on Pf. Whole-sporozoite (WSp) vaccination, targeting pre-erythrocytic (PE) parasite stages, is a promising strategy for immunization against malaria and several PfWSp-based vaccine candidates are currently undergoing clinical evaluation. In contrast, no WSp candidates have been developed for Pv, mainly due to constraints in the production of Pv sporozoites in the laboratory. Recently, we developed a novel approach for WSp vaccination against Pf based on the use of transgenic rodent P. berghei (Pb) sporozoites expressing immunogens of this human-infective parasite. We showed that this platform can be used to deliver PE Pf antigens, eliciting both targeted humoral responses and cross-species cellular immune responses against Pf. Here we explored this WSp platform for the delivery of Pv antigens. As the Pv circumsporozoite protein (CSP) is a leading vaccine candidate antigen, we generated a transgenic Pb parasite, PbviVac, that, in addition to its endogenous PbCSP, expresses PvCSP under the control of a strictly PE promoter. Immunofluorescence microscopy analyses confirmed that both the PbCSP and the PvCSP antigens are expressed in PbviVac sporozoites and liver stages and that PbviVac sporozoite infectivity of hepatic cells is similar to that of its wild-type Pb counterpart. Immunization of mice with PbviVac sporozoites elicits the production of anti-PvCSP antibodies that efficiently recognize and bind to Pv sporozoites. Our results warrant further development and evaluation of PbviVac as a surrogate for WSp vaccination against Pv malaria.
Collapse
Affiliation(s)
- Diana Moita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Teresa G Maia
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Miguel Duarte
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Carolina M Andrade
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Inês S Albuquerque
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Ankit Dwivedi
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joana C Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lilia González-Céron
- Centro Regional de Investigación en Salud Pública, Instituto Nacional de Salud Pública, Tapachula, Chiapas, México
| | - Chris J Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - António M Mendes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
8
|
Feng G, Kurtovic L, Agius PA, Aitken EH, Sacarlal J, Wines BD, Hogarth PM, Rogerson SJ, Fowkes FJI, Dobaño C, Beeson JG. Induction, decay, and determinants of functional antibodies following vaccination with the RTS,S malaria vaccine in young children. BMC Med 2022; 20:289. [PMID: 36002841 PMCID: PMC9402280 DOI: 10.1186/s12916-022-02466-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 07/06/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND RTS,S is the first malaria vaccine recommended for implementation among young children at risk. However, vaccine efficacy is modest and short-lived. Antibodies play the major role in vaccine-induced immunity, but knowledge on the induction, decay, and determinants of antibody function is limited, especially among children. Antibodies that promote opsonic phagocytosis and other cellular functions appear to be important contributors to RTS,S immunity. METHODS We studied a phase IIb trial of RTS,S/AS02 conducted in young children in malaria-endemic regions of Mozambique. We evaluated the induction of antibodies targeting the circumsporozoite protein (CSP, vaccine antigen) that interact with Fcγ-receptors (FcRγs) and promote phagocytosis (neutrophils, monocytes, THP-1 cells), antibody-dependent respiratory burst (ADRB) by neutrophils, and natural killer (NK) cell activity, as well as the temporal kinetics of responses over 5 years of follow-up (ClinicalTrials.gov registry number NCT00197041). RESULTS RTS,S vaccination induced CSP-specific IgG with FcγRIIa and FcγRIII binding activity and promoted phagocytosis by neutrophils, THP-1 monocytes, and primary human monocytes, neutrophil ADRB activity, and NK cell activation. Responses were highly heterogenous among children, and the magnitude of neutrophil phagocytosis by antibodies was relatively modest, which may reflect modest vaccine efficacy. Induction of functional antibodies was lower among children with higher malaria exposure. Functional antibody magnitude and the functional activity of antibodies largely declined within a year post-vaccination, and decay were highest in the first 6 months, consistent with the decline in vaccine efficacy over that time. Decay rates varied for different antibody parameters and decay was slower for neutrophil phagocytosis. Biostatistical modelling suggested IgG1 and IgG3 contribute in promoting FcγR binding and phagocytosis, and IgG targeting the NANP-repeat and C-terminal regions CSP were similarly important for functional activities. CONCLUSIONS Results provide new insights to understand the modest and time-limited efficacy of RTS,S in children and the induction of antibody functional activities. Improving the induction and maintenance of antibodies that promote phagocytosis and cellular functions, and combating the negative effect of malaria exposure on vaccine responses are potential strategies for improving RTS,S efficacy and longevity.
Collapse
Affiliation(s)
- Gaoqian Feng
- Burnet Institute, Melbourne, Australia.,Department of Medicine, The University of Melbourne, Melbourne, Australia
| | - Liriye Kurtovic
- Burnet Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Paul A Agius
- Burnet Institute, Melbourne, Australia.,Department of Epidemiology and Preventative Medicine, Monash University, Melbourne, Australia.,Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Elizabeth H Aitken
- Peter Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Jahit Sacarlal
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique.,Faculdade de Medicina, Universidade Eduardo Mondlane (UEM), Maputo, Mozambique
| | - Bruce D Wines
- Burnet Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia.,Department of Pathology, The University of Melbourne, Melbourne, Australia
| | - P Mark Hogarth
- Burnet Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia.,Department of Pathology, The University of Melbourne, Melbourne, Australia
| | - Stephen J Rogerson
- Department of Medicine, The University of Melbourne, Melbourne, Australia.,Peter Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Freya J I Fowkes
- Burnet Institute, Melbourne, Australia.,Department of Epidemiology and Preventative Medicine, Monash University, Melbourne, Australia.,Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Carlota Dobaño
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique.,ISGlobal, Hospital Clínic Universitat de Barcelona, Barcelona, Catalonia, Spain.,CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - James G Beeson
- Burnet Institute, Melbourne, Australia. .,Department of Medicine, The University of Melbourne, Melbourne, Australia. .,Department of Microbiology, Monash University, Clayton, Australia.
| |
Collapse
|
9
|
de Jong RM, Alkema M, Oulton T, Dumont E, Teelen K, Nakajima R, de Assis RR, Press KWD, Ngotho P, Tetteh KK, Felgner P, Marti M, Collins KA, Drakeley C, Bousema T, Stone WJ. The acquisition of humoral immune responses targeting Plasmodium falciparum sexual stages in controlled human malaria infections. Front Immunol 2022; 13:930956. [PMID: 35924245 PMCID: PMC9339717 DOI: 10.3389/fimmu.2022.930956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Individuals infected with P. falciparum develop antibody responses to intra-erythrocytic gametocyte proteins and exported gametocyte proteins present on the surface of infected erythrocytes. However, there is currently limited knowledge on the immunogenicity of gametocyte antigens and the specificity of gametocyte-induced antibody responses. In this study, we assessed antibody responses in participants of two controlled human malaria infection (CHMI) studies by ELISA, multiplexed bead-based antibody assays and protein microarray. By comparing antibody responses in participants with and without gametocyte exposure, we aimed to disentangle the antibody response induced by asexual and sexual stage parasites. We showed that after a single malaria infection, a significant anti-sexual stage humoral response is induced in malaria-naïve individuals, even after exposure to relatively low gametocyte densities (up to ~1,600 gametocytes/mL). In contrast to antibody responses to well-characterised asexual blood stage antigens that were detectable by day 21 after infection, responses to sexual stage antigens (including transmission blocking vaccine candidates Pfs48/45 and Pfs230) were only apparent at 51 days after infection. We found antigens previously associated with early gametocyte or anti-gamete immunity were highly represented among responses linked with gametocyte exposure. Our data provide detailed insights on the induction and kinetics of antibody responses to gametocytes and identify novel antigens that elicit antibody responses exclusively in individuals with gametocyte exposure. Our findings provide target identification for serological assays for surveillance of the malaria infectious reservoir, and support vaccine development by describing the antibody response to leading vaccine antigens after primary infection.
Collapse
Affiliation(s)
- Roos M. de Jong
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Manon Alkema
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Tate Oulton
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Elin Dumont
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Karina Teelen
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Rie Nakajima
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | - Rafael Ramiro de Assis
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | | | - Priscilla Ngotho
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Kevin K.A. Tetteh
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Phil Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Katharine A. Collins
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Chris Drakeley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Teun Bousema
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands,Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Will J.R. Stone
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom,*Correspondence: Will J.R. Stone,
| |
Collapse
|
10
|
Facciolà A, Visalli G, Laganà A, Di Pietro A. An Overview of Vaccine Adjuvants: Current Evidence and Future Perspectives. Vaccines (Basel) 2022; 10:vaccines10050819. [PMID: 35632575 PMCID: PMC9147349 DOI: 10.3390/vaccines10050819] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023] Open
Abstract
Vaccinations are one of the most important preventive tools against infectious diseases. Over time, many different types of vaccines have been developed concerning the antigen component. Adjuvants are essential elements that increase the efficacy of vaccination practises through many different actions, especially acting as carriers, depots, and stimulators of immune responses. For many years, few adjuvants have been included in vaccines, with aluminium salts being the most commonly used adjuvant. However, recent research has focused its attention on many different new compounds with effective adjuvant properties and improved safety. Modern technologies such as nanotechnologies and molecular biology have forcefully entered the production processes of both antigen and adjuvant components, thereby improving vaccine efficacy. Microparticles, emulsions, and immune stimulators are currently in the spotlight for their huge potential in vaccine production. Although studies have reported some potential side effects of vaccine adjuvants such as the recently recognised ASIA syndrome, the huge worth of vaccines remains unquestionable. Indeed, the recent COVID-19 pandemic has highlighted the importance of vaccines, especially in regard to managing future potential pandemics. In this field, research into adjuvants could play a leading role in the production of increasingly effective vaccines.
Collapse
Affiliation(s)
- Alessio Facciolà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
- Correspondence:
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
| | - Antonio Laganà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
- Multi-Specialist Clinical Institute for Orthopaedic Trauma Care (COT), 98124 Messina, Italy
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (G.V.); (A.L.); (A.D.P.)
| |
Collapse
|
11
|
Teixeira AR, Pérez-Cabezas B, Costa DM, Sá M, Golba S, Sefiane-Djemaoune H, Ribeiro J, Kaneko I, Iwanaga S, Yuda M, Tsuji M, Boscardin SB, Amino R, Cordeiro-da-Silva A, Tavares J. Immunization with CSP and a RIG-I Agonist is Effective in Inducing a Functional and Protective Humoral Response Against Plasmodium. Front Immunol 2022; 13:868305. [PMID: 35669785 PMCID: PMC9163323 DOI: 10.3389/fimmu.2022.868305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Malaria is a major public health concern, as a highly effective human vaccine remains elusive. The efficacy of a subunit vaccine targeting the most abundant protein of the sporozoite surface, the circumsporozoite protein (CSP) has been hindered by difficulties in generating an effective humoral response in both quantity and quality. Using the rodent Plasmodium yoelii model we report here that immunization with CSP adjuvanted with 5’ppp-dsRNA, a RIG-I agonist, confers early and long-lasting sterile protection in mice against stringent sporozoite and mosquito bite challenges. The immunization induced high levels of antibodies, which were functional in targeting and killing the sporozoites and were sustained over time through the accumulation of long-lived plasma cells in the bone marrow. Moreover, 5’ppp-dsRNA-adjuvanted immunization with the CSP of P. falciparum was also significantly protective against challenges using a transgenic PfCSP-expressing P. yoelii parasite. Conversely, using the TLR3 agonist poly(A:U) as adjuvant resulted in a formulation that despite inducing high antibody levels was unable to generate equally functional antibodies and was, consequently, less protective. In conclusion, we demonstrate that using 5’ppp-dsRNA as an adjuvant to vaccines targeting CSP induces effective anti-Plasmodium humoral immunity.
Collapse
Affiliation(s)
- Ana Rafaela Teixeira
- Host-Parasite Interactions Group, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Begoña Pérez-Cabezas
- Host-Parasite Interactions Group, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - David M. Costa
- Host-Parasite Interactions Group, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Mónica Sá
- Host-Parasite Interactions Group, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Sylvain Golba
- Center for Production and Infection of Anopheles, Institut Pasteur, Paris, France
| | | | - Joana Ribeiro
- Host-Parasite Interactions Group, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Izumi Kaneko
- Department of Medical Zoology, Mie University Graduate School of Medicine, Mie, Japan
| | - Shiroh Iwanaga
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masao Yuda
- Department of Medical Zoology, Mie University Graduate School of Medicine, Mie, Japan
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Silvia Beatriz Boscardin
- Institute for Investigation in Immunology (iii)-INCT, São Paulo, Brazil
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rogerio Amino
- Unit of Malaria Infection and Immunity, Institut Pasteur, Paris, France
| | - Anabela Cordeiro-da-Silva
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
- Parasite Disease Group, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Joana Tavares
- Host-Parasite Interactions Group, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
- *Correspondence: Joana Tavares,
| |
Collapse
|
12
|
Fabra-García A, Yang AS, Behet MC, Yap XZ, van Waardenburg Y, Kaviraj S, Lanke K, van Gemert GJ, Jore MM, Bousema T, Sauerwein RW. Human antibodies against non-circumsporozoite proteins block Plasmodium falciparum parasite development in hepatocytes. JCI Insight 2022; 7:153524. [PMID: 35167490 PMCID: PMC8986077 DOI: 10.1172/jci.insight.153524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/09/2022] [Indexed: 12/02/2022] Open
Abstract
Sporozoite-based approaches currently represent the most effective vaccine strategies for induction of sterile protection against Plasmodium falciparum (Pf) malaria. Clinical development of subunit vaccines is almost exclusively centered on the circum-sporozoite protein (CSP), an abundantly expressed protein on the sporozoite membrane. Anti-CSP antibodies are able to block sporozoite invasion and development in human hepatocytes and subsequently prevent clinical malaria. Here, we have investigated whether sporozoite-induced human antibodies with specificities different from CSP can reduce Pf-liver stage development. IgG preparations were obtained from 12 volunteers inoculated with a protective immunization regime of whole sporozoites under chloroquine prophylaxis. These IgGs were depleted for CSP specificity by affinity chromatography. Recovered non-CSP antibodies were tested for sporozoite membrane binding and for functional inhibition of sporozoite invasion of a human hepatoma cell line and hepatocytes both in vitro and in vivo. Postimmunization IgGs depleted for CS specificity of 9 of 12 donors recognized sporozoite surface antigens. Samples from 5 of 12 donors functionally reduced parasite-liver cell invasion or development using the hepatoma cell line HC-04 and FRG-huHep mice containing human liver cells. The combined data provide clear evidence that non-CSP proteins, as yet undefined, do represent antibody targets for functional immunity against Pf parasites responsible for malaria.
Collapse
Affiliation(s)
- Amanda Fabra-García
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Annie Sp Yang
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Marije C Behet
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Xi Zen Yap
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Youri van Waardenburg
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | | | - Kjerstin Lanke
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
13
|
Kurtovic L, Drew DR, Dent AE, Kazura JW, Beeson JG. Antibody Targets and Properties for Complement-Fixation Against the Circumsporozoite Protein in Malaria Immunity. Front Immunol 2021; 12:775659. [PMID: 34925347 PMCID: PMC8671933 DOI: 10.3389/fimmu.2021.775659] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/15/2021] [Indexed: 01/02/2023] Open
Abstract
The Plasmodium falciparum circumsporozoite protein (CSP) forms the basis of leading subunit malaria vaccine candidates. However, the mechanisms and specific targets of immunity are poorly defined. Recent findings suggest that antibody-mediated complement-fixation and activation play an important role in immunity. Here, we investigated the regions of CSP targeted by functional complement-fixing antibodies and the antibody properties associated with this activity. We quantified IgG, IgM, and functional complement-fixing antibody responses to different regions of CSP among Kenyan adults naturally exposed to malaria (n=102) and using a series of rabbit vaccination studies. Individuals who acquired functional complement-fixing antibodies had higher IgG, IgM and IgG1 and IgG3 to CSP. Acquired complement-fixing antibodies targeted the N-terminal, central-repeat, and C-terminal regions of CSP, and positive responders had greater antibody breadth compared to those who were negative for complement-fixing antibodies (p<0.05). Using rabbit vaccinations as a model, we confirmed that IgG specific to the central-repeat and non-repeat regions of CSP could effectively fix complement. However, vaccination with near full length CSP in rabbits poorly induced antibodies to the N-terminal region compared to naturally-acquired immunity in humans. Poor induction of N-terminal antibodies was also observed in a vaccination study performed in mice. IgG and IgM to all three regions of CSP play a role in mediating complement-fixation, which has important implications for malaria vaccine development.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Damien R. Drew
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
| | - Arlene E. Dent
- Center for Global Health and Diseases, Case Western University, Cleveland, OH, United States
| | - James W. Kazura
- Center for Global Health and Diseases, Case Western University, Cleveland, OH, United States
| | - James G. Beeson
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
14
|
Ong GH, Lian BSX, Kawasaki T, Kawai T. Exploration of Pattern Recognition Receptor Agonists as Candidate Adjuvants. Front Cell Infect Microbiol 2021; 11:745016. [PMID: 34692565 PMCID: PMC8526852 DOI: 10.3389/fcimb.2021.745016] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022] Open
Abstract
Adjuvants are used to maximize the potency of vaccines by enhancing immune reactions. Components of adjuvants include pathogen-associated molecular patterns (PAMPs) and damage-associate molecular patterns (DAMPs) that are agonists for innate immune receptors. Innate immune responses are usually activated when pathogen recognition receptors (PRRs) recognize PAMPs derived from invading pathogens or DAMPs released by host cells upon tissue damage. Activation of innate immunity by PRR agonists in adjuvants activates acquired immune responses, which is crucial to enhance immune reactions against the targeted pathogen. For example, agonists for Toll-like receptors have yielded promising results as adjuvants, which target PRR as adjuvant candidates. However, a comprehensive understanding of the type of immunological reaction against agonists for PRRs is essential to ensure the safety and reliability of vaccine adjuvants. This review provides an overview of the current progress in development of PRR agonists as vaccine adjuvants, the molecular mechanisms that underlie activation of immune responses, and the enhancement of vaccine efficacy by these potential adjuvant candidates.
Collapse
Affiliation(s)
- Guang Han Ong
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Benedict Shi Xiang Lian
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Takumi Kawasaki
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| |
Collapse
|
15
|
Mensah BA, Myers-Hansen JL, Obeng Amoako E, Opoku M, Abuaku BK, Ghansah A. Prevalence and risk factors associated with asymptomatic malaria among school children: repeated cross-sectional surveys of school children in two ecological zones in Ghana. BMC Public Health 2021; 21:1697. [PMID: 34535112 PMCID: PMC8447720 DOI: 10.1186/s12889-021-11714-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/29/2021] [Indexed: 11/27/2022] Open
Abstract
Background Asymptomatic Plasmodium infections significantly drive malaria transmission and impact control and elimination strategies, but are largely uncharacterized. We investigated the prevalence and risk factors of asymptomatic malaria infections to inform malaria control strategies in Ghana. Method Five cross-sectional surveys were conducted at the end of the peak transmission season (August–September) on 4892 school children aged between 6 and 14 years in two distinct ecological settings in Ghana between 2013 and 2017. The study sites were Begoro (forest ecology) and Cape Coast (coastal ecology). The children were screened for malaria parasites by microscopic examination of Giemsa-stained thin and thick blood films. Hemoglobin levels were measured using the Hemocue HB analyzer. In addition, height was measured and the height-for-age z-scores estimated from the reference population defined by WHO to determine children who were stunted. Proportions of categorical and means of continuous variables were compared using Chi-square test and Student’s t-test respectively, and multivariable logistic regression was done to assess risk factors associated with asymptomatic infections. Results The overall prevalence of asymptomatic malaria in the school children was higher in Begoro compared to Cape Coast (27% (95% CI: 17, 24%) vs. 24% (95% CI: 17, 24%), p value = 0.04). The study recorded three species of Plasmodium (Plasmodia falciparum, malariae, and ovale) in both sites. Plasmodium falciparum was the predominant species, accounting for about 85% of infections in both study sites. The asymptomatic school children were more likely to be anaemic (OR = 2.01, p value< 0.001) and stunted in growth (OR = 1.46, p value< 0.001). Males carried more asymptomatic infection than females (OR = 1.18, p value = 0.015). School children aged 12–14 years had more asymptomatic infections than those aged 6–8 years (OR = 1.28, p value = 0.005). Conclusion There is a considerable burden of asymptomatic malaria in the two regions of Ghana, which is associated with males, older children, anaemia, and stunted growth in children, and may have implications for malaria control and elimination strategies in Ghana.
Collapse
Affiliation(s)
- B A Mensah
- Department of Epidemiology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - J L Myers-Hansen
- Department of Epidemiology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.,Department of Parasitology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - E Obeng Amoako
- Department of Epidemiology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - M Opoku
- Department of Epidemiology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.,Department of Parasitology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - B K Abuaku
- Department of Epidemiology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - A Ghansah
- Department of Epidemiology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana. .,Department of Parasitology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.
| |
Collapse
|
16
|
TLR3 agonists: RGC100, ARNAX, and poly-IC: a comparative review. Immunol Res 2021; 69:312-322. [PMID: 34145551 PMCID: PMC8213534 DOI: 10.1007/s12026-021-09203-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/10/2021] [Indexed: 12/21/2022]
Abstract
Toll-like receptors 3 (TLR3) have been broadly studied among all TLRs over the last few decades together with its agonists due to their contribution to cancer regression. These agonists undeniably have some shared characteristics such as mimicking dsRNA but pathways through which they exhibit antitumor properties are relatively diverse. In this review, three widely studied agonists RGC100, ARNAX, and poly-IC are discussed along with their structural and physiochemical differences including the signaling cascades through which they exert their actions. Comparison has been made to identify the finest agonist with maximum effectivity and the least side effect profile.
Collapse
|
17
|
Collins KA, Brod F, Snaith R, Ulaszewska M, Longley RJ, Salman AM, Gilbert SC, Spencer AJ, Franco D, Ballou WR, Hill AVS. Ultra-low dose immunization and multi-component vaccination strategies enhance protection against malaria in mice. Sci Rep 2021; 11:10792. [PMID: 34031479 PMCID: PMC8144388 DOI: 10.1038/s41598-021-90290-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/10/2021] [Indexed: 11/10/2022] Open
Abstract
An effective vaccine would be a valuable tool for malaria control and elimination; however, the leading malaria vaccine in development, RTS,S/AS01, provided only partial protection in a Phase 3 trial. R21 is a next-generation RTS,S-like vaccine. We have previously shown in mice that R21 administered in Matrix-M is highly immunogenic, able to elicit complete protection against sporozoite challenge, and can be successfully administered with TRAP based viral-vectors resulting in enhanced protection. In this study, we developed a novel, GMP-compatible purification process for R21, and evaluated the immunogenicity and protective efficacy of ultra-low doses of both R21 and RTS,S when formulated in AS01. We demonstrated that both vaccines are highly immunogenic and also elicit comparable high levels of protection against transgenic parasites in BALB/c mice. By lowering the vaccine dose there was a trend for increased immunogenicity and sterile protection, with the highest dose vaccine groups achieving the lowest efficacy (50% sterile protection). We also evaluated the ability to combine RTS,S/AS01 with TRAP based viral-vectors and observed concurrent induction of immune responses to both antigens with minimal interference when mixing the vaccines prior to administration. These studies suggest that R21 or RTS,S could be combined with viral-vectors for a multi-component vaccination approach and indicate that low dose vaccination should be fully explored in humans to maximize potential efficacy.
Collapse
Affiliation(s)
- Katharine A Collins
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK. .,Radboud Institute for Health Science, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Florian Brod
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rebecca Snaith
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marta Ulaszewska
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rhea J Longley
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ahmed M Salman
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah C Gilbert
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexandra J Spencer
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Adrian V S Hill
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Barry A, Bradley J, Stone W, Guelbeogo MW, Lanke K, Ouedraogo A, Soulama I, Nébié I, Serme SS, Grignard L, Patterson C, Wu L, Briggs JJ, Janson O, Awandu SS, Ouedraogo M, Tarama CW, Kargougou D, Zongo S, Sirima SB, Marti M, Drakeley C, Tiono AB, Bousema T. Higher gametocyte production and mosquito infectivity in chronic compared to incident Plasmodium falciparum infections. Nat Commun 2021; 12:2443. [PMID: 33903595 PMCID: PMC8076179 DOI: 10.1038/s41467-021-22573-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 03/09/2021] [Indexed: 11/09/2022] Open
Abstract
Plasmodium falciparum gametocyte kinetics and infectivity may differ between chronic and incident infections. In the current study, we assess parasite kinetics and infectivity to mosquitoes among children (aged 5-10 years) from Burkina Faso with (a) incident infections following parasite clearance (n = 48) and (b) chronic asymptomatic infections (n = 60). In the incident infection cohort, 92% (44/48) of children develop symptoms within 35 days, compared to 23% (14/60) in the chronic cohort. All individuals with chronic infection carried gametocytes or developed them during follow-up, whereas only 35% (17/48) in the incident cohort produce gametocytes before becoming symptomatic and receiving treatment. Parasite multiplication rate (PMR) and the relative abundance of ap2-g and gexp-5 transcripts are positively associated with gametocyte production. Antibody responses are higher and PMR lower in chronic infections. The presence of symptoms and sexual stage immune responses are associated with reductions in gametocyte infectivity to mosquitoes. We observe that most incident infections require treatment before the density of mature gametocytes is sufficient to infect mosquitoes. In contrast, chronic, asymptomatic infections represent a significant source of mosquito infections. Our observations support the notion that malaria transmission reduction may be expedited by enhanced case management, involving both symptom-screening and infection detection.
Collapse
Affiliation(s)
- Aissata Barry
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
- Radboud Institute for Health Sciences and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - John Bradley
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Will Stone
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Moussa W Guelbeogo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Kjerstin Lanke
- Radboud Institute for Health Sciences and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Alphonse Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Issiaka Soulama
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Issa Nébié
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Samuel S Serme
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Lynn Grignard
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Catriona Patterson
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Lindsey Wu
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Jessica J Briggs
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Owen Janson
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Shehu S Awandu
- Radboud Institute for Health Sciences and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mireille Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Casimire W Tarama
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Désiré Kargougou
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Soumanaba Zongo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Sodiomon B Sirima
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Chris Drakeley
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Alfred B Tiono
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Teun Bousema
- Radboud Institute for Health Sciences and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands.
| |
Collapse
|
19
|
Pirahmadi S, Zakeri S, Djadid ND, Mehrizi AA. A review of combination adjuvants for malaria vaccines: a promising approach for vaccine development. Int J Parasitol 2021; 51:699-717. [PMID: 33798560 DOI: 10.1016/j.ijpara.2021.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/18/2020] [Accepted: 01/28/2021] [Indexed: 01/16/2023]
Abstract
It is obvious that there is a critical need for an efficient malaria vaccine to accelerate malaria eradication. Currently, recombinant subunit vaccination against malaria using proteins and peptides is gaining attention. However, one of the major drawbacks of this approach is the lack of an efficient and durable immune response. Therefore, subunit vaccines require adjuvants to make the vaccine sufficiently immunogenic. Considering the history of the RTS,S vaccine, it seems likely that no single adjuvant is capable of eliciting all the protective immune responses required in many malarial subunit vaccines and the use of combination adjuvants will be increasingly important as the science of malaria vaccines advances. In light of this, it appears that identifying the most effective mixture of adjuvants with minimal adverse effects offers tremendous opportunities in improving the efficacy of vaccines against malaria. Owing to the importance of a multi-adjuvanted approach in subunit malaria vaccine development, this review paper outlines some of the best known combination adjuvants used in malaria subunit vaccines, focusing on their proposed mechanisms of action, their immunological properties, and their notable results. The aim of the present review is to consolidate these findings to aid the application of these combination adjuvants in experimental malaria vaccines.
Collapse
Affiliation(s)
- Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Navid D Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Akram A Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
20
|
Kurtovic L, Wetzel D, Reiling L, Drew DR, Palmer C, Kouskousis B, Hanssen E, Wines BD, Hogarth PM, Suckow M, Jenzelewski V, Piontek M, Chan JA, Beeson JG. Novel Virus-Like Particle Vaccine Encoding the Circumsporozoite Protein of Plasmodium falciparum Is Immunogenic and Induces Functional Antibody Responses in Mice. Front Immunol 2021; 12:641421. [PMID: 33815393 PMCID: PMC8010251 DOI: 10.3389/fimmu.2021.641421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/01/2021] [Indexed: 01/10/2023] Open
Abstract
RTS,S is the leading malaria vaccine in development, but has demonstrated only moderate protective efficacy in clinical trials. RTS,S is a virus-like particle (VLP) that uses the human hepatitis B virus as scaffold to display the malaria sporozoite antigen, circumsporozoite protein (CSP). Particle formation requires four-fold excess scaffold antigen, and as a result, CSP represents only a small portion of the final vaccine construct. Alternative VLP or nanoparticle platforms that reduce the amount of scaffold antigen and increase the amount of the target CSP antigen present in particles may enhance vaccine immunogenicity and efficacy. Here, we describe the production and characterization of a novel VLP that uses the small surface antigen (dS) of duck hepatitis B virus to display CSP. The CSP-dS fusion protein successfully formed VLPs without the need for excess scaffold antigen, and thus CSP represented a larger portion of the vaccine construct. CSP-dS formed large particles approximately 31-74 nm in size and were confirmed to display CSP on the surface. CSP-dS VLPs were highly immunogenic in mice and induced antibodies to multiple regions of CSP, even when administered at a lower vaccine dosage. Vaccine-induced antibodies demonstrated relevant functional activities, including Fc-dependent interactions with complement and Fcγ-receptors, previously identified as important in malaria immunity. Further, vaccine-induced antibodies had similar properties (epitope-specificity and avidity) to monoclonal antibodies that are protective in mouse models. Our novel platform to produce VLPs without excess scaffold protein has wide implications for the future development of vaccines for malaria and other infectious diseases.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Departments of Immunology and Pathology and Infectious Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | | | - Linda Reiling
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
| | - Damien R. Drew
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
| | | | | | - Eric Hanssen
- The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Bruce D. Wines
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Departments of Immunology and Pathology and Infectious Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - P. Mark Hogarth
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Departments of Immunology and Pathology and Infectious Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | | | | | | | - Jo-Anne Chan
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Departments of Immunology and Pathology and Infectious Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - James G. Beeson
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Departments of Immunology and Pathology and Infectious Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
21
|
A chemokine-fusion vaccine targeting immature dendritic cells elicits elevated antibody responses to malaria sporozoites in infant macaques. Sci Rep 2021; 11:1220. [PMID: 33441615 PMCID: PMC7807052 DOI: 10.1038/s41598-020-79427-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 11/02/2020] [Indexed: 11/18/2022] Open
Abstract
Infants and young children are the groups at greatest risk for severe disease resulting from Plasmodium falciparum infection. We previously demonstrated in mice that a protein vaccine composed of the chemokine macrophage inflammatory protein 3α genetically fused to the minimally truncated circumsporozoite protein of P. falciparum (MCSP) elicits high concentrations of specific antibody and significant reduction of liver sporozoite load in a mouse model system. In the current study, a squalene based adjuvant (AddaVax, InvivoGen, San Diego, Ca) equivalent to the clinically approved MF59 (Seqiris, Maidenhead, UK) elicited greater antibody responses in mice than the previously employed adjuvant polyinosinic:polycytidylic acid, ((poly(I:C), InvivoGen, San Diego, Ca) and the clinically approved Aluminum hydroxide gel (Alum, Invivogen, San Diego, Ca) adjuvant. Use of the AddaVax adjuvant also expanded the range of IgG subtypes elicited by mouse vaccination. Sera passively transferred into mice from MCSP/AddaVax immunized 1 and 6 month old macaques significantly reduced liver sporozoite load upon sporozoite challenge. Protective antibody concentrations attained by passive transfer in the mice were equivalent to those observed in infant macaques 18 weeks after the final immunization. The efficacy of this vaccine in a relevant non-human primate model indicates its potential usefulness for the analogous high risk human population.
Collapse
|
22
|
Pfeifer K, Ergal İ, Koller M, Basen M, Schuster B, Rittmann SKMR. Archaea Biotechnology. Biotechnol Adv 2020; 47:107668. [PMID: 33271237 DOI: 10.1016/j.biotechadv.2020.107668] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022]
Abstract
Archaea are a domain of prokaryotic organisms with intriguing physiological characteristics and ecological importance. In Microbial Biotechnology, archaea are historically overshadowed by bacteria and eukaryotes in terms of public awareness, industrial application, and scientific studies, although their biochemical and physiological properties show a vast potential for a wide range of biotechnological applications. Today, the majority of microbial cell factories utilized for the production of value-added and high value compounds on an industrial scale are bacterial, fungal or algae based. Nevertheless, archaea are becoming ever more relevant for biotechnology as their cultivation and genetic systems improve. Some of the main advantages of archaeal cell factories are the ability to cultivate many of these often extremophilic organisms under non-sterile conditions, and to utilize inexpensive feedstocks often toxic to other microorganisms, thus drastically reducing cultivation costs. Currently, the only commercially available products of archaeal cell factories are bacterioruberin, squalene, bacteriorhodopsin and diether-/tetraether-lipids, all of which are produced utilizing halophiles. Other archaeal products, such as carotenoids and biohydrogen, as well as polyhydroxyalkanoates and methane are in early to advanced development stages, respectively. The aim of this review is to provide an overview of the current state of Archaea Biotechnology by describing the actual state of research and development as well as the industrial utilization of archaeal cell factories, their role and their potential in the future of sustainable bioprocessing, and to illustrate their physiological and biotechnological potential.
Collapse
Affiliation(s)
- Kevin Pfeifer
- Archaea Physiology & Biotechnology Group, Department of Functional and Evolutionary Ecology, Universität Wien, Wien, Austria; Institute of Synthetic Bioarchitectures, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, Wien, Austria
| | - İpek Ergal
- Archaea Physiology & Biotechnology Group, Department of Functional and Evolutionary Ecology, Universität Wien, Wien, Austria
| | - Martin Koller
- Office of Research Management and Service, c/o Institute of Chemistry, University of Graz, Austria
| | - Mirko Basen
- Microbial Physiology Group, Division of Microbiology, Institute of Biological Sciences, University of Rostock, Rostock, Germany
| | - Bernhard Schuster
- Institute of Synthetic Bioarchitectures, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, Wien, Austria
| | - Simon K-M R Rittmann
- Archaea Physiology & Biotechnology Group, Department of Functional and Evolutionary Ecology, Universität Wien, Wien, Austria.
| |
Collapse
|
23
|
Micoli F, Alfini R, Di Benedetto R, Necchi F, Schiavo F, Mancini F, Carducci M, Palmieri E, Balocchi C, Gasperini G, Brunelli B, Costantino P, Adamo R, Piccioli D, Saul A. GMMA Is a Versatile Platform to Design Effective Multivalent Combination Vaccines. Vaccines (Basel) 2020; 8:E540. [PMID: 32957610 PMCID: PMC7564227 DOI: 10.3390/vaccines8030540] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 01/21/2023] Open
Abstract
Technology platforms are an important strategy to facilitate the design, development and implementation of vaccines to combat high-burden diseases that are still a threat for human populations, especially in low- and middle-income countries, and to address the increasing number and global distribution of pathogens resistant to antimicrobial drugs. Generalized Modules for Membrane Antigens (GMMA), outer membrane vesicles derived from engineered Gram-negative bacteria, represent an attractive technology to design affordable vaccines. Here, we show that GMMA, decorated with heterologous polysaccharide or protein antigens, leads to a strong and effective antigen-specific humoral immune response in mice. Importantly, GMMA promote enhanced immunogenicity compared to traditional formulations (e.g., recombinant proteins and glycoconjugate vaccines), without negative impact to the anti-GMMA immune response. Our findings support the use of GMMA as a "plug and play" technology for the development of effective combination vaccines targeting different bugs at the same time.
Collapse
Affiliation(s)
- Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Roberta Di Benedetto
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Francesca Necchi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Fabiola Schiavo
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Francesca Mancini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Martina Carducci
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | | | - Gianmarco Gasperini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| | | | | | - Roberto Adamo
- GSK, 53100 Siena, Italy; (C.B.); (B.B.); (P.C.); (R.A.); (D.P.)
| | - Diego Piccioli
- GSK, 53100 Siena, Italy; (C.B.); (B.B.); (P.C.); (R.A.); (D.P.)
| | - Allan Saul
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., 53100 Siena, Italy; (R.A.); (R.D.B.); (F.N.); (F.S.); (F.M.); (M.C.); (E.P.); (G.G.); (A.S.)
| |
Collapse
|
24
|
A Multistage Formulation Based on Full-Length CSP and AMA-1 Ectodomain of Plasmodium vivax Induces High Antibody Titers and T-cells and Partially Protects Mice Challenged with a Transgenic Plasmodium berghei Parasite. Microorganisms 2020; 8:microorganisms8060916. [PMID: 32560380 PMCID: PMC7356588 DOI: 10.3390/microorganisms8060916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/13/2020] [Accepted: 06/14/2020] [Indexed: 01/23/2023] Open
Abstract
Infections with Plasmodium vivax are predominant in the Americas, representing 75% of malaria cases. Previously perceived as benign, malaria vivax is, in fact, a highly debilitating and economically important disease. Considering the high complexity of the malaria parasite life cycle, it has been hypothesized that an effective vaccine formulation against Plasmodium should contain multiple antigens expressed in different parasite stages. Based on that, we analyzed a recombinant P. vivax vaccine formulation mixing the apical membrane antigen 1 ectodomain (PvAMA-1) and a full-length circumsporozoite protein (PvCSP-AllFL) previously studied by our group, which elicits a potent antibody response in mice. Genetically distinct strains of mice (C57BL/6 and BALB/c) were immunized with the proteins, alone or in combination, in the presence of poly(I:C) adjuvant, a TLR3 agonist. In C57BL/6, high-antibody titers were induced against PvAMA-1 and the three PvCSP variants (VK210, VK247, and P. vivax-like). Meanwhile, mixing PvAMA-1 with PvCSP-AllFL had no impact on total IgG antibody titers, which were long-lasting. Moreover, antibodies from immunized mice recognized VK210 sporozoites and blood-stage parasites by immunofluorescence assay. However, in the BALB/c model, the antibody response against PvCSP-AllFL was relatively low. PvAMA-1-specific CD3+CD4+ and CD3+CD8+ T-cell responses were observed in C57BL/6 mice, and the cellular response was impaired by PvCSP-AllFL combination. More relevant, the multistage vaccine formulation provided partial protection in mice challenged with a transgenic Plasmodium berghei sporozoite expressing the homologous PvCSP protein.
Collapse
|
25
|
van den Hoogen LL, Stresman G, Présumé J, Romilus I, Mondélus G, Elismé T, Existe A, Hamre KES, Ashton RA, Druetz T, Joseph V, Beeson JG, Singh SK, Boncy J, Eisele TP, Chang MA, Lemoine JF, Tetteh KKA, Rogier E, Drakeley C. Selection of Antibody Responses Associated With Plasmodium falciparum Infections in the Context of Malaria Elimination. Front Immunol 2020; 11:928. [PMID: 32499783 PMCID: PMC7243477 DOI: 10.3389/fimmu.2020.00928] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/21/2020] [Indexed: 12/30/2022] Open
Abstract
In our aim to eliminate malaria, more sensitive tools to detect residual transmission are quickly becoming essential. Antimalarial antibody responses persist in the blood after a malaria infection and provide a wider window to detect exposure to infection compared to parasite detection metrics. Here, we aimed to select antibody responses associated with recent and cumulative exposure to malaria using cross-sectional survey data from Haiti, an elimination setting. Using a multiplex bead assay, we generated data for antibody responses (immunoglobulin G) to 23 Plasmodium falciparum targets in 29,481 participants across three surveys. This included one community-based survey in which participants were enrolled during household visits and two sentinel group surveys in which participants were enrolled at schools and health facilities. First, we correlated continuous antibody responses with age (Spearman) to determine which showed strong age-related associations indicating accumulation over time with limited loss. AMA-1 and MSP-119 antibody levels showed the strongest correlation with age (0.47 and 0.43, p < 0.001) in the community-based survey, which was most representative of the underlying age structure of the population, thus seropositivity to either of these antibodies was considered representative of cumulative exposure to malaria. Next, in the absence of a gold standard for recent exposure, we included antibody responses to the remaining targets to predict highly sensitive rapid diagnostic test (hsRDT) status using receiver operating characteristic curves. For this, only data from the survey with the highest hsRDT prevalence was used (7.2%; 348/4,849). The performance of the top two antigens in the training dataset (two-thirds of the dataset; n = 3,204)-Etramp 5 ag 1 and GLURP-R0 (area-under-the-curve, AUC, 0.892 and 0.825, respectively)-was confirmed in the test dataset (remaining one-third of the dataset; n = 1,652, AUC 0.903 and 0.848, respectively). As no further improvement was seen by combining seropositivity to GLURP-R0 and Etramp 5 ag 1 (p = 0.266), seropositivity to Etramp 5 ag 1 alone was selected as representative of current or recent exposure to malaria. The validation of antibody responses associated with these exposure histories simplifies analyses and interpretation of antibody data and facilitates the application of results to evaluate programs.
Collapse
Affiliation(s)
- Lotus L. van den Hoogen
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA, United States
| | - Gillian Stresman
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | | | - Gina Mondélus
- Laboratoire National de Santé Publique, Port-au-Prince, Haiti
| | - Tamara Elismé
- Laboratoire National de Santé Publique, Port-au-Prince, Haiti
| | | | - Karen E. S. Hamre
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, United States
- CDC Foundation, Atlanta, GA, United States
| | - Ruth A. Ashton
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA, United States
| | - Thomas Druetz
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA, United States
- Department of Social and Preventive Medicine, University of Montreal School of Public Health, Montreal, QC, Canada
| | - Vena Joseph
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA, United States
| | - James G. Beeson
- Burnet Institute, Melbourne, VIC, Australia
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
- Central Clinical School and Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Susheel K. Singh
- Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Department of Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark
| | - Jacques Boncy
- Laboratoire National de Santé Publique, Port-au-Prince, Haiti
| | - Thomas P. Eisele
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA, United States
| | - Michelle A. Chang
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jean F. Lemoine
- Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti
| | - Kevin K. A. Tetteh
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Eric Rogier
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Chris Drakeley
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
26
|
Oyen D, Torres JL, Aoto PC, Flores-Garcia Y, Binter Š, Pholcharee T, Carroll S, Reponen S, Wash R, Liang Q, Lemiale F, Locke E, Bradley A, King CR, Emerling D, Kellam P, Zavala F, Ward AB, Wilson IA. Structure and mechanism of monoclonal antibody binding to the junctional epitope of Plasmodium falciparum circumsporozoite protein. PLoS Pathog 2020; 16:e1008373. [PMID: 32150583 PMCID: PMC7082059 DOI: 10.1371/journal.ppat.1008373] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/19/2020] [Accepted: 01/31/2020] [Indexed: 11/21/2022] Open
Abstract
Lasting protection has long been a goal for malaria vaccines. The major surface antigen on Plasmodium falciparum sporozoites, the circumsporozoite protein (PfCSP), has been an attractive target for vaccine development and most protective antibodies studied to date interact with the central NANP repeat region of PfCSP. However, it remains unclear what structural and functional characteristics correlate with better protection by one antibody over another. Binding to the junctional region between the N-terminal domain and central NANP repeats has been proposed to result in superior protection: this region initiates with the only NPDP sequence followed immediately by NANP. Here, we isolated antibodies in Kymab mice immunized with full-length recombinant PfCSP and two protective antibodies were selected for further study with reactivity against the junctional region. X-ray and EM structures of two monoclonal antibodies, mAb667 and mAb668, shed light on their differential affinity and specificity for the junctional region. Importantly, these antibodies also bind to the NANP repeat region with equal or better affinity. A comparison with an NANP-only binding antibody (mAb317) revealed roughly similar but statistically distinct levels of protection against sporozoite challenge in mouse liver burden models, suggesting that junctional antibody protection might relate to the ability to also cross-react with the NANP repeat region. Our findings indicate that additional efforts are necessary to isolate a true junctional antibody with no or much reduced affinity to the NANP region to elucidate the role of the junctional epitope in protection. The circumsporozoite protein (CSP) of Plasmodium falciparum malaria has been the foundation for the design of transmission blocking malaria vaccines. To date, the most promising CSP-based vaccine candidate is RTS,S, which consists of the central repeating NANP amino-acid sequence and the C-terminal domain of CSP fused to hepatitis B surface antigen that assembles into virus-like particles. Potential shortcomings of RTS,S includes the lack of other potential CSP epitopes such as the junctional epitope, which is located between the N-terminal domain of CSP and the start of the NANP repeat region. Here, we elicited antibodies against full-length CSP and screened for junctional epitope binding. We then used an array of biophysical techniques to elucidate the nature of the binding and tested the level of two protective antibodies in a mouse challenge model. Although the antibodies were able to bind both junctional and NANP epitopes, the in vivo data showed distinct levels of protection between themselves and also to an NANP-only binder. Our data suggest that their protection ability may be related to the strong cross-reactivity with NANP epitopes. Since all reported junctional antibodies to date have dual-specificity, we suggest that a true junctional binder with no or very low NANP affinity, if one can be found, is essential to evaluate the contribution of the junctional epitope to protection.
Collapse
Affiliation(s)
- David Oyen
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jonathan L. Torres
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Phillip C. Aoto
- Department of Pharmacology, University of California at San Diego, La Jolla, California, United States of America
| | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Špela Binter
- Kymab Ltd., The Bennet Building (B930), Babraham Research Campus, Cambridge, United Kingdom
| | - Tossapol Pholcharee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Sean Carroll
- Atreca Inc., South San Francisco, California, United States of America
| | - Sini Reponen
- Atreca Inc., South San Francisco, California, United States of America
| | - Rachael Wash
- Kymab Ltd., The Bennet Building (B930), Babraham Research Campus, Cambridge, United Kingdom
| | - Qi Liang
- Kymab Ltd., The Bennet Building (B930), Babraham Research Campus, Cambridge, United Kingdom
| | - Franck Lemiale
- PATH’s Malaria Vaccine Initiative, PATH Center for Vaccine Innovation and Access, Washington, United States of America
| | - Emily Locke
- PATH’s Malaria Vaccine Initiative, PATH Center for Vaccine Innovation and Access, Washington, United States of America
| | - Allan Bradley
- Kymab Ltd., The Bennet Building (B930), Babraham Research Campus, Cambridge, United Kingdom
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - C. Richter King
- PATH’s Malaria Vaccine Initiative, PATH Center for Vaccine Innovation and Access, Washington, United States of America
| | - Daniel Emerling
- Atreca Inc., South San Francisco, California, United States of America
| | - Paul Kellam
- Kymab Ltd., The Bennet Building (B930), Babraham Research Campus, Cambridge, United Kingdom
- Department of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
27
|
Amelia F, Iyori M, Emran TB, Yamamoto DS, Genshi K, Otsuka H, Onoue Y, Yusuf Y, Islam A, Yoshida S. Down-selecting circumsporozoite protein-based malaria vaccine: A comparison of malaria sporozoite challenge model. Parasite Immunol 2020; 41:e12624. [PMID: 30883819 DOI: 10.1111/pim.12624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/28/2019] [Accepted: 03/08/2019] [Indexed: 12/18/2022]
Abstract
Plasmodium falciparum circumsporozoite protein (PfCSP) is the main target antigen in development of pre-erythrocytic malaria vaccines. To evaluate PfCSP vaccines in animal models, challenge by intravenous sporozoite injection is preferentially used. However, in clinical trials, vaccinated human volunteers are exposed to the bites of malaria-infected mosquitoes. In this study, we down-selected Escherichia coli-produced full-length PfCSP (PfCSP-F) and its three truncated PfCSPs based on their abilities to elicit immune response and protection in mice against two challenge models. We showed that immunization with three doses of PfCSP-F elicited high anti-PfCSP antibody titres and 100% protection against the bites of infected mosquitoes. Meanwhile, three-dose truncated PfCSP induced 60%-70% protection after immunization with each truncated PfCSP. Heterologous prime-boost immunization regimen with adenovirus-PfCSP-F and R32LR greatly induced complete protection against intravenous sporozoite injection. Our results suggest that Abs to both anti-repeat and anti-nonrepeat regions induced by PfCSP-F are required to confer complete protection against challenge by the bites of infected mosquitoes, whereas anti-repeat Abs play an important role in protection against intravenous sporozoite injection. Our findings provide a potential clinical application that PfCSP-F vaccine induces potent Abs capable of neutralizing sporozoites in the dermis inoculated by infected mosquitoes and subsequently sporozoites in the blood circulation.
Collapse
Affiliation(s)
- Fitri Amelia
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan.,Department of Chemistry, Universitas Negeri Padang, Padang, Indonesia
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Talha Bin Emran
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke, Japan
| | - Kento Genshi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Hiromu Otsuka
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Yutaro Onoue
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Ashekul Islam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| |
Collapse
|
28
|
van den Hoogen LL, Présumé J, Romilus I, Mondélus G, Elismé T, Sepúlveda N, Stresman G, Druetz T, Ashton RA, Joseph V, Eisele TP, Hamre KES, Chang MA, Lemoine JF, Tetteh KKA, Boncy J, Existe A, Drakeley C, Rogier E. Quality control of multiplex antibody detection in samples from large-scale surveys: the example of malaria in Haiti. Sci Rep 2020; 10:1135. [PMID: 31980693 PMCID: PMC6981173 DOI: 10.1038/s41598-020-57876-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022] Open
Abstract
Measuring antimalarial antibodies can estimate transmission in a population. To compare outputs, standardized laboratory testing is required. Here we describe the in-country establishment and quality control (QC) of a multiplex bead assay (MBA) for three sero-surveys in Haiti. Total IgG data against 21 antigens were collected for 32,758 participants. Titration curves of hyperimmune sera were included on assay plates, assay signals underwent 5-parameter regression, and inspection of the median and interquartile range (IQR) for the y-inflection point was used to determine assay precision. The medians and IQRs were similar for Surveys 1 and 2 for most antigens, while the IQRs increased for some antigens in Survey 3. Levey-Jennings charts for selected antigens provided a pass/fail criterion for each assay plate and, of 387 assay plates, 13 (3.4%) were repeated. Individual samples failed if IgG binding to the generic glutathione-S-transferase protein was observed, with 659 (2.0%) samples failing. An additional 455 (1.4%) observations failed due to low bead numbers (<20/analyte). The final dataset included 609,438 anti-malaria IgG data points from 32,099 participants; 96.6% of all potential data points if no QC failures had occurred. The MBA can be deployed with high-throughput data collection and low inter-plate variability while ensuring data quality.
Collapse
Affiliation(s)
- Lotus L van den Hoogen
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK.
| | | | | | - Gina Mondélus
- Laboratoire National de Santé Publique, Port-au-Prince, Haiti
| | - Tamara Elismé
- Laboratoire National de Santé Publique, Port-au-Prince, Haiti
| | - Nuno Sepúlveda
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
- Centre of Statistics and Applications, University of Lisbon, Lisbon, Portugal
| | - Gillian Stresman
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
| | - Thomas Druetz
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health & Tropical Medicine, New Orleans, Louisiana, USA
- Department of Social and Preventive Medicine, University of Montreal School of Public Health, Montreal, Canada
| | - Ruth A Ashton
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health & Tropical Medicine, New Orleans, Louisiana, USA
| | - Vena Joseph
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health & Tropical Medicine, New Orleans, Louisiana, USA
| | - Thomas P Eisele
- Center for Applied Malaria Research and Evaluation, Tulane University School of Public Health & Tropical Medicine, New Orleans, Louisiana, USA
| | - Karen E S Hamre
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- CDC Foundation, Atlanta, Georgia, USA
| | - Michelle A Chang
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jean F Lemoine
- Ministère de la santé publique et de la population, Port-au-Prince, Haiti
| | - Kevin K A Tetteh
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
| | - Jacques Boncy
- Laboratoire National de Santé Publique, Port-au-Prince, Haiti
| | | | - Chris Drakeley
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
| | - Eric Rogier
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
29
|
Heide J, Wildner NH, Ackermann C, Wittner M, Marget M, Sette A, Sidney J, Jacobs T, Schulze Zur Wiesch J. Detection of EXP1-Specific CD4+ T Cell Responses Directed Against a Broad Range of Epitopes Including Two Promiscuous MHC Class II Binders During Acute Plasmodium falciparum Malaria. Front Immunol 2020; 10:3037. [PMID: 32038611 PMCID: PMC6993587 DOI: 10.3389/fimmu.2019.03037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/11/2019] [Indexed: 01/02/2023] Open
Abstract
Background: T cells are thought to play a major role in conferring immunity against malaria. This study aimed to comprehensively define the breadth and specificity of the Plasmodium falciparum (P. falciparum)-specific CD4+ T cell response directed against the exported protein 1 (EXP1) in a cohort of patients diagnosed with acute malaria. Methods: Peripheral blood mononuclear cells of 44 patients acutely infected with P. falciparum, and of one patient infected with P. vivax, were stimulated and cultured in vitro with an overlapping set of 31 P. falciparum-specific 13-17-mer peptides covering the entire EXP1 sequence. EXP1-specific T cell responses were analyzed by ELISPOT and intracellular cytokine staining for interferon-γ production after re-stimulation with individual peptides. For further characterization of the epitopes, in silico and in vitro human leukocyte antigen (HLA) binding studies and fine mapping assays were performed. Results: We detected one or more EXP1-specific CD4+ T cell responses (mean: 1.09, range 0–5) in 47% (21/45) of our patients. Responses were directed against 15 of the 31 EXP1 peptides. Peptides EXP1-P13 (aa60-74) and P15 (aa70-85) were detected by 18% (n = 8) and 27% (n = 12) of the 45 patients screened. The optimal length, as well as the corresponding most likely HLA-restriction, of each of these two peptides was assessed. Interestingly, we also identified one CD4+ T cell response against peptide EXP1-P15 in a patient who was infected with P. vivax but not falciparum. Conclusions: This first detailed characterization of novel EXP1-specific T cell epitopes provides important information for future analysis with major histocompatibility complex-multimer technology as well as for immunomonitoring and vaccine design.
Collapse
Affiliation(s)
- Janna Heide
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Nils H Wildner
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christin Ackermann
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Wittner
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Matthias Marget
- Department of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States.,Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Thomas Jacobs
- Protozoa Immunology, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | - Julian Schulze Zur Wiesch
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
30
|
Singh SK, Plieskatt J, Chourasia BK, Singh V, Bolscher JM, Dechering KJ, Adu B, López-Méndez B, Kaviraj S, Locke E, King CR, Theisen M. The Plasmodium falciparum circumsporozoite protein produced in Lactococcus lactis is pure and stable. J Biol Chem 2019; 295:403-414. [PMID: 31792057 DOI: 10.1074/jbc.ra119.011268] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/03/2019] [Indexed: 12/20/2022] Open
Abstract
The Plasmodium falciparum circumsporozoite protein (PfCSP) is a sporozoite surface protein whose role in sporozoite motility and cell invasion has made it the leading candidate for a pre-erythrocytic malaria vaccine. However, production of high yields of soluble recombinant PfCSP, including its extensive NANP and NVDP repeats, has proven problematic. Here, we report on the development and characterization of a secreted, soluble, and stable full-length PfCSP (containing 4 NVDP and 38 NANP repeats) produced in the Lactococcus lactis expression system. The recombinant full-length PfCSP, denoted PfCSP4/38, was produced initially with a histidine tag and purified by a simple two-step procedure. Importantly, the recombinant PfCSP4/38 retained a conformational epitope for antibodies as confirmed by both in vivo and in vitro characterizations. We characterized this complex protein by HPLC, light scattering, MS analysis, differential scanning fluorimetry, CD, SDS-PAGE, and immunoblotting with conformation-dependent and -independent mAbs, which confirmed it to be both pure and soluble. Moreover, we found that the recombinant protein is stable at both frozen and elevated-temperature storage conditions. When we used L. lactis-derived PfCSP4/38 to immunize mice, it elicited high levels of functional antibodies that had the capacity to modify sporozoite motility in vitro We concluded that the reported yield, purity, results of biophysical analyses, and stability of PfCSP4/38 warrant further consideration of using the L. lactis system for the production of circumsporozoite proteins for preclinical and clinical applications in malaria vaccine development.
Collapse
Affiliation(s)
- Susheel K Singh
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | | | - Bishwanath Kumar Chourasia
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Vandana Singh
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | | | | | - Bright Adu
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Blanca López-Méndez
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | - Emily Locke
- PATH's Malaria Vaccine Initiative, Washington, D. C. 20001
| | - C Richter King
- PATH's Malaria Vaccine Initiative, Washington, D. C. 20001
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark.
| |
Collapse
|
31
|
Pattinson DJ, Apte SH, Wibowo N, Chuan YP, Rivera-Hernandez T, Groves PL, Lua LH, Middelberg APJ, Doolan DL. Chimeric Murine Polyomavirus Virus-Like Particles Induce Plasmodium Antigen-Specific CD8 + T Cell and Antibody Responses. Front Cell Infect Microbiol 2019; 9:215. [PMID: 31275867 PMCID: PMC6593135 DOI: 10.3389/fcimb.2019.00215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/03/2019] [Indexed: 12/28/2022] Open
Abstract
An effective vaccine against the Plasmodium parasite is likely to require the induction of robust antibody and T cell responses. Chimeric virus-like particles are an effective vaccine platform for induction of antibody responses, but their capacity to induce robust cellular responses and cell-mediated protection against pathogen challenge has not been established. To evaluate this, we produced chimeric constructs using the murine polyomavirus structural protein with surface-exposed CD8+ or CD4+ T cell or B cell repeat epitopes derived from the Plasmodium yoelii circumsporozoite protein, and assessed immunogenicity and protective capacity in a murine model. Robust CD8+ T cell responses were induced by immunization with the chimeric CD8+ T cell epitope virus-like particles, however CD4+ T cell responses were very low. The B cell chimeric construct induced robust antibody responses but there was no apparent synergy when T cell and B cell constructs were administered as a pool. A heterologous prime/boost regimen using plasmid DNA priming followed by a VLP boost was more effective than homologous VLP immunization for cellular immunity and protection. These data show that chimeric murine polyomavirus virus-like particles are a good platform for induction of CD8+ T cell responses as well as antibody responses.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan
- Antibody Formation/immunology
- Antigens, Protozoan/immunology
- B-Lymphocytes
- CD4-Positive T-Lymphocytes
- CD8-Positive T-Lymphocytes/immunology
- Disease Models, Animal
- Epitopes, B-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/immunology
- Immunity, Cellular
- Immunization
- Immunization, Secondary
- Malaria Vaccines
- Mice
- Mice, Inbred BALB C
- Plasmodium yoelii
- Polyomavirus/genetics
- Polyomavirus/immunology
- Protozoan Proteins/immunology
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- David J. Pattinson
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Simon H. Apte
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nani Wibowo
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Yap P. Chuan
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Tania Rivera-Hernandez
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Penny L. Groves
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Linda H. Lua
- Protein Expression Facility, University of Queensland, Brisbane, QLD, Australia
| | - Anton P. J. Middelberg
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Denise L. Doolan
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
32
|
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
|
33
|
Kobayashi T, Jain A, Liang L, Obiero JM, Hamapumbu H, Stevenson JC, Thuma PE, Lupiya J, Chaponda M, Mulenga M, Mamini E, Mharakurwa S, Gwanzura L, Munyati S, Mutambu S, Felgner P, Davies DH, Moss WJ. Distinct Antibody Signatures Associated with Different Malaria Transmission Intensities in Zambia and Zimbabwe. mSphere 2019; 4:e00061-19. [PMID: 30918058 PMCID: PMC6437277 DOI: 10.1128/mspheredirect.00061-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/30/2022] Open
Abstract
Antibodies to Plasmodium falciparum are specific biomarkers that can be used to monitor parasite exposure over broader time frames than microscopy, rapid diagnostic tests, or molecular assays. Consequently, seroprevalence surveys can assist with monitoring the impact of malaria control interventions, particularly in the final stages of elimination, when parasite incidence is low. The protein array format to measure antibodies to diverse P. falciparum antigens requires only small sample volumes and is high throughput, permitting the monitoring of malaria transmission on large spatial and temporal scales. We expanded the use of a protein microarray to assess malaria transmission in settings beyond those with a low malaria incidence. Antibody responses in children and adults were profiled, using a P. falciparum protein microarray, through community-based surveys in three areas in Zambia and Zimbabwe at different stages of malaria control and elimination. These three epidemiological settings had distinct serological profiles reflective of their malaria transmission histories. While there was little correlation between transmission intensity and antibody signals (magnitude or breadth) in adults, there was a clear correlation in children younger than 5 years of age. Antibodies in adults appeared to be durable even in the absence of significant recent transmission, whereas antibodies in children provided a more accurate picture of recent levels of transmission intensity. Seroprevalence studies in children could provide a valuable marker of progress toward malaria elimination.IMPORTANCE As malaria approaches elimination in many areas of the world, monitoring the effect of control measures becomes more important but challenging. Low-level infections may go undetected by conventional tests that depend on parasitemia, particularly in immune individuals, who typically show no symptoms of malaria. In contrast, antibodies persist after parasitemia and may provide a more accurate picture of recent exposure. Only a few parasite antigens-mainly vaccine candidates-have been evaluated in seroepidemiological studies. We examined antibody responses to 500 different malaria proteins in blood samples collected through community-based surveillance from areas with low, medium, and high malaria transmission intensities. The breadth of the antibody responses in adults was broad in all three settings and was a poor correlate of recent exposure. In contrast, children represented a better sentinel population for monitoring recent malaria transmission. These data will help inform the use of multiplex serology for malaria surveillance.
Collapse
Affiliation(s)
- Tamaki Kobayashi
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Aarti Jain
- Vaccine Research & Development Center, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Li Liang
- Vaccine Research & Development Center, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Joshua M Obiero
- Vaccine Research & Development Center, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | | | - Jennifer C Stevenson
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Macha Research Trust, Choma, Zambia
| | - Philip E Thuma
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Macha Research Trust, Choma, Zambia
| | - James Lupiya
- Tropical Diseases Research Centre, Ndola, Zambia
| | | | | | - Edmore Mamini
- Biomedical Research and Training Institute, Harare, Zimbabwe
| | | | | | - Shungu Munyati
- Biomedical Research and Training Institute, Harare, Zimbabwe
| | - Susan Mutambu
- National Institute of Health Research, Harare, Zimbabwe
| | - Philip Felgner
- Vaccine Research & Development Center, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - D Huw Davies
- Vaccine Research & Development Center, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - William J Moss
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
34
|
van den Hoogen LL, Walk J, Oulton T, Reuling IJ, Reiling L, Beeson JG, Coppel RL, Singh SK, Draper SJ, Bousema T, Drakeley C, Sauerwein R, Tetteh KKA. Antibody Responses to Antigenic Targets of Recent Exposure Are Associated With Low-Density Parasitemia in Controlled Human Plasmodium falciparum Infections. Front Microbiol 2019; 9:3300. [PMID: 30700984 PMCID: PMC6343524 DOI: 10.3389/fmicb.2018.03300] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 12/18/2018] [Indexed: 12/05/2022] Open
Abstract
The majority of malaria infections in low transmission settings remain undetectable by conventional diagnostics. A powerful model to identify antibody responses that allow accurate detection of recent exposure to low-density infections is controlled human malaria infection (CHMI) studies in which healthy volunteers are infected with the Plasmodium parasite. We aimed to evaluate antibody responses in malaria-naïve volunteers exposed to a single CHMI using a custom-made protein microarray. All participants developed a blood-stage infection with peak parasite densities up to 100 parasites/μl in the majority of participants (50/54), while the remaining four participants had peak densities between 100 and 200 parasites/μl. There was a strong correlation between parasite density and antibody responses associated with the most reactive blood-stage targets 1 month after CHMI (Etramp 5, GLURP-R2, MSP4 and MSP1-19; Spearman’s ρ = 0.82, p < 0.001). Most volunteers developed antibodies against a potential marker of recent exposure: Etramp 5 (37/45, 82%). Our findings justify validation in endemic populations to define a minimum set of antigens needed to detect exposure to natural low-density infections.
Collapse
Affiliation(s)
- Lotus L van den Hoogen
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jona Walk
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Tate Oulton
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Isaie J Reuling
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia.,Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia.,Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Ross L Coppel
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Susheel K Singh
- Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Department of International Health, Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark
| | - Simon J Draper
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Chris Drakeley
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Robert Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Kevin K A Tetteh
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
35
|
Identification of Immune Signatures of Novel Adjuvant Formulations Using Machine Learning. Sci Rep 2018; 8:17508. [PMID: 30504893 PMCID: PMC6269591 DOI: 10.1038/s41598-018-35452-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/06/2018] [Indexed: 11/27/2022] Open
Abstract
Adjuvants have long been critical components of vaccines, but the exact mechanisms of their action and precisely how they alter or enhance vaccine-induced immune responses are often unclear. In this study, we used broad immunoprofiling of antibody, cellular, and cytokine responses, combined with data integration and machine learning to gain insight into the impact of different adjuvant formulations on vaccine-induced immune responses. A Self-Assembling Protein Nanoparticles (SAPN) presenting the malarial circumsporozoite protein (CSP) was used as a model vaccine, adjuvanted with three different liposomal formulations: liposome plus Alum (ALFA), liposome plus QS21 (ALFQ), and both (ALFQA). Using a computational approach to integrate the immunoprofiling data, we identified distinct vaccine-induced immune responses and developed a multivariate model that could predict the adjuvant condition from immune response data alone with 92% accuracy (p = 0.003). The data integration also revealed that commonly used readouts (i.e. serology, frequency of T cells producing IFN-γ, IL2, TNFα) missed important differences between adjuvants. In summary, broad immune-profiling in combination with machine learning methods enabled the reliable and clear definition of immune signatures for different adjuvant formulations, providing a means for quantitatively characterizing the complex roles that adjuvants can play in vaccine-induced immunity. The approach described here provides a powerful tool for identifying potential immune correlates of protection, a prerequisite for the rational pairing of vaccines candidates and adjuvants.
Collapse
|
36
|
Ubillos I, Jiménez A, Vidal M, Bowyer PW, Gaur D, Dutta S, Gamain B, Coppel R, Chauhan V, Lanar D, Chitnis C, Angov E, Beeson J, Cavanagh D, Campo JJ, Aguilar R, Dobaño C. Optimization of incubation conditions of Plasmodium falciparum antibody multiplex assays to measure IgG, IgG 1-4, IgM and IgE using standard and customized reference pools for sero-epidemiological and vaccine studies. Malar J 2018; 17:219. [PMID: 29859096 PMCID: PMC5984756 DOI: 10.1186/s12936-018-2369-3] [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: 02/12/2018] [Accepted: 05/28/2018] [Indexed: 12/15/2022] Open
Abstract
Background The quantitative suspension array technology (qSAT) is a useful platform for malaria immune marker discovery. However, a major challenge for large sero-epidemiological and malaria vaccine studies is the comparability across laboratories, which requires the access to standardized control reagents for assay optimization, to monitor performance and improve reproducibility. Here, the Plasmodium falciparum antibody reactivities of the newly available WHO reference reagent for anti-malaria human plasma (10/198) and of additional customized positive controls were examined with seven in-house qSAT multiplex assays measuring IgG, IgG1–4 subclasses, IgM and IgE against a panel of 40 antigens. The different positive controls were tested at different incubation times and temperatures (4 °C overnight, 37 °C 2 h, room temperature 1 h) to select the optimal conditions. Results Overall, the WHO reference reagent had low IgG2, IgG4, IgM and IgE, and also low anti-CSP antibody levels, thus this reagent was enriched with plasmas from RTS,S-vaccinated volunteers to be used as standard for CSP-based vaccine studies. For the IgM assay, another customized plasma pool prepared with samples from malaria primo-infected adults with adequate IgM levels proved to be more adequate as a positive control. The range and magnitude of IgG and IgG1–4 responses were highest when the WHO reference reagent was incubated with antigen-coupled beads at 4 °C overnight. IgG levels measured in the negative control did not vary between incubations at 37 °C 2 h and 4 °C overnight, indicating no difference in unspecific binding. Conclusions With this study, the immunogenicity profile of the WHO reference reagent, including seven immunoglobulin isotypes and subclasses, and more P. falciparum antigens, also those included in the leading RTS,S malaria vaccine, was better characterized. Overall, incubation of samples at 4 °C overnight rendered the best performance for antibody measurements against the antigens tested. Although the WHO reference reagent performed well to measure IgG to the majority of the common P. falciparum blood stage antigens tested, customized pools may need to be used as positive controls depending on the antigens (e.g. pre-erythrocytic proteins of low natural immunogenicity) and isotypes/subclasses (e.g. IgM) under study. Electronic supplementary material The online version of this article (10.1186/s12936-018-2369-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Itziar Ubillos
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Paul W Bowyer
- Bacteriology Division, MHRA-NIBSC, South Mimms, Potter Bars, EN6 3QG, UK
| | - Deepak Gaur
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.,Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sheetij Dutta
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Benoit Gamain
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge UMR_S1134, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Ross Coppel
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Virander Chauhan
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - David Lanar
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Chetan Chitnis
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Evelina Angov
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - James Beeson
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, Australia
| | - David Cavanagh
- Institute of Immunology & Infection Research and Centre for Immunity, Infection & Evolution, Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, King's Buildings, Charlotte Auerbach Rd, Edinburgh, EH9 3FL, UK
| | - Joseph J Campo
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.
| |
Collapse
|
37
|
Prime-boost vaccination with recombinant protein and adenovirus-vector expressing Plasmodium vivax circumsporozoite protein (CSP) partially protects mice against Pb/Pv sporozoite challenge. Sci Rep 2018; 8:1118. [PMID: 29348479 PMCID: PMC5773670 DOI: 10.1038/s41598-017-19063-6] [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: 03/30/2017] [Accepted: 12/21/2017] [Indexed: 12/20/2022] Open
Abstract
Vaccine development against Plasmodium vivax malaria lags behind that for Plasmodium falciparum. To narrow this gap, we administered recombinant antigens based on P. vivax circumsporozoite protein (CSP) to mice. We expressed in Pichia pastoris two chimeric proteins by merging the three central repeat regions of different CSP alleles (VK210, VK247, and P. vivax-like). The first construct (yPvCSP-AllFL) contained the fused repeat regions flanked by N- and C-terminal regions. The second construct (yPvCSP-AllCT) contained the fused repeat regions and the C-terminal domain, plus RI region. Mice were vaccinated with three doses of yPvCSP in adjuvants Poly (I:C) or Montanide ISA720. We also used replication-defective adenovirus vectors expressing CSP of human serotype 5 (AdHu5) and chimpanzee serotype 68 (AdC68) for priming mice which were subsequently boosted twice with yPvCSP proteins in Poly (I:C) adjuvant. Regardless of the regime used, immunized mice generated high IgG titres specific to all CSP alleles. After challenge with P. berghei ANKA transgenic parasites expressing Pb/PvVK210 or Pb/PvVK247 sporozoites, significant time delays for parasitemia were observed in all vaccinated mice. These vaccine formulations should be clinically tried for their potential as protective universal vaccine against P. vivax malaria.
Collapse
|
38
|
Jin Y, He X, Andoh‐Kumi K, Fraser RZ, Lu M, Goodman RE. Evaluating Potential Risks of Food Allergy and Toxicity of Soy Leghemoglobin Expressed in Pichia pastoris. Mol Nutr Food Res 2018; 62:1700297. [PMID: 28921896 PMCID: PMC5813221 DOI: 10.1002/mnfr.201700297] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/09/2017] [Indexed: 11/24/2022]
Abstract
SCOPE The Soybean (Glycine max) leghemoglobin c2 (LegHb) gene was introduced into Pichia pastoris yeast for sustainable production of a heme-carrying protein, for organoleptic use in plant-based meat. The potential allergenicity and toxicity of LegHb and 17 Pichia host-proteins each representing ≥1% of total protein in production batches are evaluated by literature review, bioinformatics sequence comparisons to known allergens or toxins, and in vitro pepsin digestion. METHODS AND RESULTS Literature searches found no evidence of allergenicity or toxicity for these proteins. There are no significant sequence matches of LegHb to known allergens or toxins. Eleven Pichia proteins have modest identity matches to minor environmental allergens and 13 Pichia proteins have significant matches to proteins from toxic sources. Yet the matched allergens and toxins have similar matches to proteins from the commonly consumed yeast Saccharomyces cerevisiae, without evidence of food allergy or toxicity. The demonstrated history of safe use indicates additional tests for allergenicity and toxicity are not needed. The LegHb and Pichia sp. proteins were rapidly digested by pepsin at pH 2. CONCLUSION These results demonstrate that foods containing recombinant soy LegHb produced in Pichia sp. are unlikely to present an unacceptable risk of allergenicity or toxicity to consumers.
Collapse
Affiliation(s)
- Yuan Jin
- Food Allergy Research and Resource ProgramDept. of Food Science & TechnologyUniversity of Nebraska‐LincolnLincolnNEUSA
| | - Xiaoyun He
- College of Food Science and Nutritional EngineeringChina Agricultural UniversityBeijingChina
| | - Kwame Andoh‐Kumi
- Food Allergy Research and Resource ProgramDept. of Food Science & TechnologyUniversity of Nebraska‐LincolnLincolnNEUSA
| | | | - Mei Lu
- Food Allergy Research and Resource ProgramDept. of Food Science & TechnologyUniversity of Nebraska‐LincolnLincolnNEUSA
| | - Richard E. Goodman
- Food Allergy Research and Resource ProgramDept. of Food Science & TechnologyUniversity of Nebraska‐LincolnLincolnNEUSA
| |
Collapse
|
39
|
Walk J, Reuling IJ, Behet MC, Meerstein-Kessel L, Graumans W, van Gemert GJ, Siebelink-Stoter R, van de Vegte-Bolmer M, Janssen T, Teelen K, de Wilt JHW, de Mast Q, van der Ven AJ, Diez Benavente E, Campino S, Clark TG, Huynen MA, Hermsen CC, Bijker EM, Scholzen A, Sauerwein RW. Modest heterologous protection after Plasmodium falciparum sporozoite immunization: a double-blind randomized controlled clinical trial. BMC Med 2017; 15:168. [PMID: 28903777 PMCID: PMC5598044 DOI: 10.1186/s12916-017-0923-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/02/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND A highly efficacious vaccine is needed for malaria control and eradication. Immunization with Plasmodium falciparum NF54 parasites under chemoprophylaxis (chemoprophylaxis and sporozoite (CPS)-immunization) induces the most efficient long-lasting protection against a homologous parasite. However, parasite genetic diversity is a major hurdle for protection against heterologous strains. METHODS We conducted a double-blind, randomized controlled trial in 39 healthy participants of NF54-CPS immunization by bites of 45 NF54-infected (n = 24 volunteers) or uninfected mosquitoes (placebo; n = 15 volunteers) against a controlled human malaria infection with the homologous NF54 or the genetically distinct NF135.C10 and NF166.C8 clones. Cellular and humoral immune assays were performed as well as genetic characterization of the parasite clones. RESULTS NF54-CPS immunization induced complete protection in 5/5 volunteers against NF54 challenge infection at 14 weeks post-immunization, but sterilely protected only 2/10 and 1/9 volunteers against NF135.C10 and NF166.C8 challenge infection, respectively. Post-immunization plasma showed a significantly lower capacity to block heterologous parasite development in primary human hepatocytes compared to NF54. Whole genome sequencing showed that NF135.C10 and NF166.C8 have amino acid changes in multiple antigens targeted by CPS-induced antibodies. Volunteers protected against heterologous challenge were among the stronger immune responders to in vitro parasite stimulation. CONCLUSIONS Although highly protective against homologous parasites, NF54-CPS-induced immunity is less effective against heterologous parasite clones both in vivo and in vitro. Our data indicate that whole sporozoite-based vaccine approaches require more potent immune responses for heterologous protection. TRIAL REGISTRATION This trial is registered in clinicaltrials.gov, under identifier NCT02098590 .
Collapse
Affiliation(s)
- Jona Walk
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands
| | - Isaie J Reuling
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands
| | - Marije C Behet
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands
| | - Lisette Meerstein-Kessel
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands.,Radboud Institute of Molecular Life Sciences and Center for Molecular and Biomolecular Informatics, Radboud University Medical Center, Geert Grooteplein 28, CMBI 260, 6500 HB, Nijmegen, The Netherlands
| | - Wouter Graumans
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands
| | - Rianne Siebelink-Stoter
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands
| | - Marga van de Vegte-Bolmer
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands
| | - Thorsten Janssen
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands
| | - Karina Teelen
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands
| | - Johannes H W de Wilt
- Department of Surgery, Radboud University Medical Center, Geert Grooteplein 10, Surgery 618, 6500 HB, Nijmegen, The Netherlands
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein 10, Internal Medicine 456, 6500 HB, Nijmegen, The Netherlands
| | - André J van der Ven
- Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein 10, Internal Medicine 456, 6500 HB, Nijmegen, The Netherlands
| | - Ernest Diez Benavente
- London School of Hygiene and Tropical Medicine, Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London, WC1E 7HT, UK
| | - Susana Campino
- London School of Hygiene and Tropical Medicine, Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London, WC1E 7HT, UK
| | - Taane G Clark
- London School of Hygiene and Tropical Medicine, Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London, WC1E 7HT, UK.,London School of Hygiene and Tropical Medicine, Department of Infectious Disease Epidemiology, Faculty of Infectious and Tropical Diseases, London, WC1E 7HT, UK
| | - Martijn A Huynen
- Radboud Institute of Molecular Life Sciences and Center for Molecular and Biomolecular Informatics, Radboud University Medical Center, Geert Grooteplein 28, CMBI 260, 6500 HB, Nijmegen, The Netherlands
| | - Cornelus C Hermsen
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands
| | - Else M Bijker
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands.,Present Address: Department of Pediatrics, Radboud university medical center, Geert Grooteplein 10, Pediatrics 804, 6500 HB, Nijmegen, The Netherlands
| | - Anja Scholzen
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands.,Present Address: Innatoss Laboratories B.V., Kloosterstraat 9, RE3124, 5349 AB, Oss, The Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein 28, Microbiology 268, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
40
|
Genito CJ, Beck Z, Phares TW, Kalle F, Limbach KJ, Stefaniak ME, Patterson NB, Bergmann-Leitner ES, Waters NC, Matyas GR, Alving CR, Dutta S. Liposomes containing monophosphoryl lipid A and QS-21 serve as an effective adjuvant for soluble circumsporozoite protein malaria vaccine FMP013. Vaccine 2017; 35:3865-3874. [DOI: 10.1016/j.vaccine.2017.05.070] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 05/03/2017] [Accepted: 05/24/2017] [Indexed: 11/24/2022]
|
41
|
Collins KA, Snaith R, Cottingham MG, Gilbert SC, Hill AVS. Enhancing protective immunity to malaria with a highly immunogenic virus-like particle vaccine. Sci Rep 2017; 7:46621. [PMID: 28422178 PMCID: PMC5395940 DOI: 10.1038/srep46621] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/03/2017] [Indexed: 12/11/2022] Open
Abstract
The leading malaria vaccine in development is the circumsporozoite protein (CSP)-based particle vaccine, RTS,S, which targets the pre-erythrocytic stage of Plasmodium falciparum infection. It induces modest levels of protective efficacy, thought to be mediated primarily by CSP-specific antibodies. We aimed to enhance vaccine efficacy by generating a more immunogenic CSP-based particle vaccine and therefore developed a next-generation RTS,S-like vaccine, called R21. The major improvement is that in contrast to RTS,S, R21 particles are formed from a single CSP-hepatitis B surface antigen (HBsAg) fusion protein, and this leads to a vaccine composed of a much higher proportion of CSP than in RTS,S. We demonstrate that in BALB/c mice R21 is immunogenic at very low doses and when administered with the adjuvants Abisco-100 and Matrix-M it elicits sterile protection against transgenic sporozoite challenge. Concurrent induction of potent cellular and humoral immune responses was also achieved by combining R21 with TRAP-based viral vectors and protective efficacy was significantly enhanced. In addition, in contrast to RTS,S, only a minimal antibody response to the HBsAg carrier was induced. These studies identify an anti-sporozoite vaccine component that may improve upon the current leading malaria vaccine RTS,S. R21 is now under evaluation in Phase 1/2a clinical trials.
Collapse
Affiliation(s)
- Katharine A. Collins
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Rebecca Snaith
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Matthew G. Cottingham
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Sarah C. Gilbert
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Adrian V. S. Hill
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| |
Collapse
|
42
|
Espinosa DA, Christensen D, Muñoz C, Singh S, Locke E, Andersen P, Zavala F. Robust antibody and CD8 + T-cell responses induced by P. falciparum CSP adsorbed to cationic liposomal adjuvant CAF09 confer sterilizing immunity against experimental rodent malaria infection. NPJ Vaccines 2017; 2. [PMID: 28936360 PMCID: PMC5603302 DOI: 10.1038/s41541-017-0011-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite several decades of extensive research, the development of a highly efficacious malaria vaccine has yet to be accomplished. While the RTS,S malaria vaccine candidate shows the potential to prevent a substantial number of clinical malaria cases, significant improvements in protective efficacy are still needed. Multiple studies have shown that RTS,S induces protective antibody and CD4+ T-cell responses, but limited or negligible CD8+ T cells. In this study, we evaluated the immunogenicity and protective capacity of full-length recombinant Plasmodium falciparum circumsporozoite protein administered with the novel cationic liposomal adjuvant system CAF09. Using newly developed transgenic rodent malaria parasites expressing the full-length Plasmodium falciparum circumsporozoite protein, we demonstrate that this liposome-based protein-in-adjuvant formulation is capable of inducing robust antibody and CD8+ T-cell responses that strongly inhibit parasite infection and development of liver stages, conferring durable sterilizing immunity. These findings underscore the potential of liposome-based adjuvants for inducing robust humoral and CD8+ T-cell responses and warrant further studies toward the development of novel subunit vaccine formulations with this adjuvant system. A vaccine consisting of parasitic proteins enveloped by fatty molecules provides comprehensive protection against malaria in a rodent model, Previous and current malaria vaccines concentrate on priming antibodies to recognize malarial infection, despite evidence that, by activating ‘killer’ CD8+ T cells, greater protection is conferred against the disease. Fidel Zavala, of the Johns Hopkins University, United States, and an international group of researchers developed their vaccine by encapsulating proteins from the malaria-causing parasite Plasmodium falciparum in fat-based carriers called liposomes. In past experiments, killer T cells recruited via this vaccine-type have effectively protected against other diseases. In this study, the vaccine induced both CD8+ T cell and antibody responses and provided significant immunity against P. falciparum-instigated malaria. As a highly efficacious vaccine against malaria is not yet available, this research will likely prove invaluable in guiding further studies.
Collapse
Affiliation(s)
- Diego A Espinosa
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Christian Muñoz
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Emily Locke
- PATH Malaria Vaccine Initiative, Washington DC, USA
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Phares TW, May AD, Genito CJ, Hoyt NA, Khan FA, Porter MD, DeBot M, Waters NC, Saudan P, Dutta S. Rhesus macaque and mouse models for down-selecting circumsporozoite protein based malaria vaccines differ significantly in immunogenicity and functional outcomes. Malar J 2017; 16:115. [PMID: 28288639 PMCID: PMC5347822 DOI: 10.1186/s12936-017-1766-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/28/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Non-human primates, such as the rhesus macaques, are the preferred model for down-selecting human malaria vaccine formulations, but the rhesus model is expensive and does not allow for direct efficacy testing of human malaria vaccines. Transgenic rodent parasites expressing genes of human Plasmodium are now routinely used for efficacy studies of human malaria vaccines. Mice have however rarely predicted success in human malaria trials and there is scepticism whether mouse studies alone are sufficient to move a vaccine candidate into the clinic. METHODS A comparison of immunogenicity, fine-specificity and functional activity of two Alum-adjuvanted Plasmodium falciparum circumsporozoite protein (CSP)-based vaccines was conducted in mouse and rhesus models. One vaccine was a soluble recombinant protein (CSP) and the other was the same CSP covalently conjugated to the Qβ phage particle (Qβ-CSP). RESULTS Mice showed different kinetics of antibody responses and different sensitivity to the NANP-repeat and N-terminal epitopes as compared to rhesus. While mice failed to discern differences between the protective efficacy of CSP versus Qβ-CSP vaccine following direct challenge with transgenic Plasmodium berghei parasites, rhesus serum from the Qβ-CSP-vaccinated animals induced higher in vivo sporozoite neutralization activity. CONCLUSIONS Despite some immunologic parallels between models, these data demonstrate that differences between the immune responses induced in the two models risk conflicting decisions regarding potential vaccine utility in humans. In combination with historical observations, the data presented here suggest that although murine models may be useful for some purposes, non-human primate models may be more likely to predict the human response to investigational vaccines.
Collapse
Affiliation(s)
- Timothy W Phares
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Anthony D May
- Division of Veterinary Medicine, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Christopher J Genito
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Nathan A Hoyt
- Division of Veterinary Medicine, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Farhat A Khan
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Michael D Porter
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Margot DeBot
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Norman C Waters
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Philippe Saudan
- Cytos Biotechnology, Wagistrasse 25, 8952, Schlieren, Switzerland
| | - Sheetij Dutta
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA.
| |
Collapse
|
44
|
Janitzek CM, Matondo S, Thrane S, Nielsen MA, Kavishe R, Mwakalinga SB, Theander TG, Salanti A, Sander AF. Bacterial superglue generates a full-length circumsporozoite protein virus-like particle vaccine capable of inducing high and durable antibody responses. Malar J 2016; 15:545. [PMID: 27825348 PMCID: PMC5101663 DOI: 10.1186/s12936-016-1574-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 10/27/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Malaria, caused by Plasmodium falciparum, continues to have a devastating impact on global health, emphasizing the great need for a malaria vaccine. The circumsporozoite protein (CSP) is an attractive target for a malaria vaccine, and forms a major component of RTS,S, the most clinically advanced malaria vaccine. The clinical efficacy of RTS,S has been moderate, yet has demonstrated the viability of a CSP-based malaria vaccine. In this study, a vaccine comprised of the full-length CSP antigen presented on a virus-like particle (VLP) is produced using a split-intein conjugation system (SpyTag/SpyCatcher) and the immunogenicity is tested in mice. METHODS Full-length 3d7 CSP protein was genetically fused at the C-terminus to SpyCatcher. The CSP-SpyCatcher antigen was then covalently attached (via the SpyTag/SpyCatcher interaction) to Acinetobacter phage AP205 VLPs which were modified to display one SpyTag per VLP subunit. To evaluate the VLP-display effect, the immunogenicity of the VLP vaccine was tested in mice and compared to a control vaccine containing AP205 VLPs plus unconjugated CSP. RESULTS Full-length CSP was conjugated at high density (an average of 112 CSP molecules per VLP) to AP205 SpyTag-VLPs. Vaccination of mice with the CSP Spy-VLP vaccine resulted in significantly increased antibody titres over a course of 7 months as compared to the control group (2.6-fold higher at 7 months after immunization). Furthermore, the CSP Spy-VLP vaccine appears to stimulate production of IgG2a antibodies, which has been linked with a more efficient clearing of intracellular parasite infection. CONCLUSION This study demonstrates that the high-density display of CSP on SpyTag-VLPs, significantly increases the level and quality of the vaccine-induced humoral response, compared to a control vaccine consisting of soluble CSP plus AP205 VLPs. The SpyTag-VLP platform utilized in this study constitutes a versatile and rapid method to develop highly immunogenic vaccines. It might serve as a generic tool for the cost-effective development of effective VLP-vaccines, e.g., against malaria.
Collapse
Affiliation(s)
- Christoph M Janitzek
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sungwa Matondo
- Kilimanjaro Christian Medical University College (KCMUCo), Moshi, Tanzania
| | - Susan Thrane
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Morten A Nielsen
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Reginald Kavishe
- Kilimanjaro Christian Medical University College (KCMUCo), Moshi, Tanzania
| | - Steve B Mwakalinga
- Kilimanjaro Christian Medical University College (KCMUCo), Moshi, Tanzania
| | - Thor G Theander
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Ali Salanti
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark. .,Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Adam F Sander
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark. .,Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark.
| |
Collapse
|
45
|
Mitchell RA, Altszuler R, Frevert U, Nardin EH. Skin scarification with Plasmodium falciparum peptide vaccine using synthetic TLR agonists as adjuvants elicits malaria sporozoite neutralizing immunity. Sci Rep 2016; 6:32575. [PMID: 27624667 PMCID: PMC5021941 DOI: 10.1038/srep32575] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 08/10/2016] [Indexed: 12/19/2022] Open
Abstract
Malaria eradication will require a combination of vector control, chemotherapy and an easily administered vaccine. Sterile immunity can be elicited in humans by immunization with sporozoites, the infective stage injected by bite of the mosquito vector, however, whole parasite vaccines present formidable logistical challenges for production, storage and administration. The “gold standard” for infectious disease eradiation, the Smallpox Eradication Programme, utilized mass immunization using the skin scarification (SS) route. SS may more closely mimic the natural route of malaria infection initiated by sporozoites injected by mosquito bite which elicits both neutralizing antibodies and protective cell mediated immunity. We investigated the potential of SS immunization using a malaria repeat peptide containing a protective B cell epitope of Plasmodium falciparum, the most lethal human species, and delivery vehicles containing TLR agonists as adjuvants. In a murine model, SS immunization with peptide in combination with TLR-7/8 and -9 agonists elicited high levels of systemic sporozoite neutralizing antibody, Th1- type CD4+ T cells and resistance to challenge by bites of infected mosquitoes. SS provides the potential to elicit humoral immunity to target Plasmodium at multiple stages of its complex life cycle.
Collapse
Affiliation(s)
- Robert A Mitchell
- Department of Microbiology, Division of Parasitology, New York University School of Medicine, New York, NY, USA
| | - Rita Altszuler
- Department of Microbiology, Division of Parasitology, New York University School of Medicine, New York, NY, USA
| | - Ute Frevert
- Department of Microbiology, Division of Parasitology, New York University School of Medicine, New York, NY, USA
| | - Elizabeth H Nardin
- Department of Microbiology, Division of Parasitology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
46
|
Identification of GLA/SE as an effective adjuvant for the induction of robust humoral and cell-mediated immune responses to EBV-gp350 in mice and rabbits. Vaccine 2016; 34:2562-9. [PMID: 27085175 DOI: 10.1016/j.vaccine.2016.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 04/02/2016] [Accepted: 04/05/2016] [Indexed: 02/07/2023]
Abstract
Childhood infection with Epstein-Barr virus (EBV) is often asymptomatic and may result in mild flu-like symptoms, but exposure during adolescence and young adulthood can lead to acute infectious mononucleosis (AIM) with a pathology characterized by swollen lymph nodes, sore throat, and severe fatigue lasting weeks or months. A vaccine targeting the envelope glycoprotein gp350 adjuvanted with aluminum hydroxide complexed with the TLR4 agonist monophosphoryl lipid A (MPLA) achieved a 78% reduction in AIM incidence in a small phase II trial of college-age individuals, but development of this vaccine was halted by the manufacturer. Here, we report the evaluation in mice and rabbits of an EBV-gp350 vaccine combined with an adjuvant composed of the synthetic TLR4 agonist glucopyranosyl lipid A (GLA) integrated into stable emulsion (SE). In mice, GLA/SE-adjuvanted gp350 generated high IgG titers (both IgG1 and IgG2a/c subtypes), elevated EBV-neutralizing antibody titers, and robust poly-functional anti-gp350 CD4(+) T cell responses. In addition, GLA/SE routinely demonstrated superior performance over aluminum hydroxide in all immunological readouts, including induction of durable neutralizing antibody titers out to at least 1 year post-vaccination. Both components of the GLA/SE adjuvant were found to be required to get optimal responses in both arms of the immune response: specifically, SE for neutralizing antibodies and GLA for induction of T cell responses. Furthermore, this vaccine also elicited high neutralizing antibody titers in a second species, rabbit. These promising results suggest that clinical development of a vaccine comprised of EBV-gp350 plus GLA/SE has the potential to prevent AIM in post-adolescents.
Collapse
|
47
|
Martins KAO, Cooper CL, Stronsky SM, Norris SLW, Kwilas SA, Steffens JT, Benko JG, van Tongeren SA, Bavari S. Adjuvant-enhanced CD4 T Cell Responses are Critical to Durable Vaccine Immunity. EBioMedicine 2015; 3:67-78. [PMID: 26870818 PMCID: PMC4739439 DOI: 10.1016/j.ebiom.2015.11.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 01/08/2023] Open
Abstract
Protein-based vaccines offer a safer alternative to live-attenuated or inactivated vaccines but have limited immunogenicity. The identification of adjuvants that augment immunogenicity, specifically in a manner that is durable and antigen-specific, is therefore critical for advanced development. In this study, we use the filovirus virus-like particle (VLP) as a model protein-based vaccine in order to evaluate the impact of four candidate vaccine adjuvants on enhancing long term protection from Ebola virus challenge. Adjuvants tested include poly-ICLC (Hiltonol), MPLA, CpG 2395, and alhydrogel. We compared and contrasted antibody responses, neutralizing antibody responses, effector T cell responses, and T follicular helper (Tfh) cell frequencies with each adjuvant's impact on durable protection. We demonstrate that in this system, the most effective adjuvant elicits a Th1-skewed antibody response and strong CD4 T cell responses, including an increase in Tfh frequency. Using immune-deficient animals and adoptive transfer of serum and cells from vaccinated animals into naïve animals, we further demonstrate that serum and CD4 T cells play a critical role in conferring protection within effective vaccination regimens. These studies inform on the requirements of long term immune protection, which can potentially be used to guide screening of clinical-grade adjuvants for vaccine clinical development. Adjuvants can prolong the protection afforded by protein-based vaccines and impact adaptive immune responses Enhanced CD4 T cell responses, helper and effector, correlate with duration of protection Durable protection from ma-EBOV is associated with Tfh frequency, Th1 antibody titers, and effector CD4 T cells
Protein-based vaccines are extremely safe, but they sometimes require the addition of adjuvants to enhance immunogenicity. In this study, we compared the impact of multiple adjuvants on immunogenicity, focusing on the duration of vaccine-mediated protection in mice. We then looked at how each adjuvant impacted the immune response in order to identify correlates of that long lasting immunity. The most effective adjuvant/vaccine combinations elicited multifunctional CD4 T cell responses and a Th1-skewed antibody response. By transferring antigen-experienced CD4 T cells and serum into naïve animals, we demonstrated that both CD4 T cells and serum were critical for durable vaccine-mediated protection.
Collapse
Key Words
- Adjuvant
- BME, beta mercaptoethanol
- CD, cluster of differentiation
- DSCF, Dwass, Steel, Critchlow-Fligner
- Durable protection
- ELISA, Enzyme linked immunosorbent assay
- ELISPOT, enzyme-linked immunospot assay
- Ebola virus
- FACS, fluorescence activated cell sorting
- FBS, fetal bovine serum
- GP, glycoprotein
- IACUC, Institutional Animal Care and Use Committee
- IM, intramuscular
- IP, intraperitoneal
- IQR, interquartile range
- Immune correlates
- LN, lymph node
- MPLA, monophosphoryl lipid A
- NAb, neutralizing antibody
- Ns, not significant
- PBS, phosphate buffered saline
- PRR, pattern recognition receptor
- Pfu, plaque forming unit
- PsVNA, pseudovirion neutralization assay
- TLR, Toll-like receptor
- USAMRIID, United States Army Medical Research Institute of Infectious Diseases
- VLP, virus-like particle
- Vaccine
- ma-EBOV, mouse-adapted Ebola virus
Collapse
Affiliation(s)
- Karen A O Martins
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Christopher L Cooper
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sabrina M Stronsky
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sarah L W Norris
- Research Support Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Steven A Kwilas
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Jesse T Steffens
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Jacqueline G Benko
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sean A van Tongeren
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA
| | - Sina Bavari
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, USA.
| |
Collapse
|
48
|
Parra M, Liu X, Derrick SC, Yang A, Molina-Cruz A, Barillas-Mury C, Zheng H, Thao Pham P, Sedegah M, Belmonte A, Litilit DD, Waldmann TA, Kumar S, Morris SL, Perera LP. Co-expression of Interleukin-15 Enhances the Protective Immune Responses Induced by Immunization with a Murine Malaria MVA-Based Vaccine Encoding the Circumsporozoite Protein. PLoS One 2015; 10:e0141141. [PMID: 26505634 PMCID: PMC4624717 DOI: 10.1371/journal.pone.0141141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/04/2015] [Indexed: 01/01/2023] Open
Abstract
Malaria remains a major global public health problem with an estimated 200 million cases detected in 2012. Although the most advanced candidate malaria vaccine (RTS,S) has shown promise in clinical trials, its modest efficacy and durability have created uncertainty about the impact of RTS,S immunization (when used alone) on global malaria transmission. Here we describe the development and characterization of a novel modified vaccinia virus Ankara (MVA)–based malaria vaccine which co-expresses the Plasmodium yoelii circumsporozoite protein (CSP) and IL-15. Vaccination/challenge studies showed that C57BL/6 mice immunized with the MVA-CSP/IL15 vaccine were protected significantly better against a P. yoelii 17XNL sporozoite challenge than either mice immunized with an MVA vaccine expressing only CSP or naïve controls. Importantly, the levels of total anti-CSP IgG were elevated about 100-fold for the MVA-CSP/IL15 immunized group compared to mice immunized with the MVA-CSP construct that does not express IL-15. Among the IgG subtypes, the IL-15 expressing MVA-CSP vaccine induced levels of IgG1 (8 fold) and IgG2b (80 fold) higher than the MVA-CSP construct. The significantly enhanced humoral responses and protection detected after immunization with the MVA-CSP/IL15 vaccine suggest that this IL-15 expressing MVA construct could be considered in the development of future malaria immunization strategies.
Collapse
Affiliation(s)
- Marcela Parra
- Food and Drug Administration, Center for Biologics Evaluation and Research, Silver Spring, MD, 20993, United States of America
| | - Xia Liu
- Food and Drug Administration, Center for Biologics Evaluation and Research, Silver Spring, MD, 20993, United States of America
| | - Steven C. Derrick
- Food and Drug Administration, Center for Biologics Evaluation and Research, Silver Spring, MD, 20993, United States of America
| | - Amy Yang
- Food and Drug Administration, Center for Biologics Evaluation and Research, Silver Spring, MD, 20993, United States of America
| | - Alvaro Molina-Cruz
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, 20852, United States of America
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, 20852, United States of America
| | - Hong Zheng
- Food and Drug Administration, Center for Biologics Evaluation and Research, Silver Spring, MD, 20993, United States of America
| | - Phuong Thao Pham
- Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
| | - Martha Sedegah
- Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
| | - Arnel Belmonte
- Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
| | - Dianne D. Litilit
- Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
| | - Thomas A. Waldmann
- National Cancer Institute, Bethesda, MD, 20892, United States of America
| | - Sanjai Kumar
- Food and Drug Administration, Center for Biologics Evaluation and Research, Silver Spring, MD, 20993, United States of America
| | - Sheldon L. Morris
- Food and Drug Administration, Center for Biologics Evaluation and Research, Silver Spring, MD, 20993, United States of America
| | - Liyanage P. Perera
- National Cancer Institute, Bethesda, MD, 20892, United States of America
- * E-mail:
| |
Collapse
|
49
|
Reversible Conformational Change in the Plasmodium falciparum Circumsporozoite Protein Masks Its Adhesion Domains. Infect Immun 2015; 83:3771-80. [PMID: 26169272 DOI: 10.1128/iai.02676-14] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 07/02/2015] [Indexed: 11/20/2022] Open
Abstract
The extended rod-like Plasmodium falciparum circumsporozoite protein (CSP) is comprised of three primary domains: a charged N terminus that binds heparan sulfate proteoglycans, a central NANP repeat domain, and a C terminus containing a thrombospondin-like type I repeat (TSR) domain. Only the last two domains are incorporated in RTS,S, the leading malaria vaccine in phase 3 trials that, to date, protects about 50% of vaccinated children against clinical disease. A seroepidemiological study indicated that the N-terminal domain might improve the efficacy of a new CSP vaccine. Using a panel of CSP-specific monoclonal antibodies, well-characterized recombinant CSPs, label-free quantitative proteomics, and in vitro inhibition of sporozoite invasion, we show that native CSP is N-terminally processed in the mosquito host and undergoes a reversible conformational change to mask some epitopes in the N- and C-terminal domains until the sporozoite interacts with the liver hepatocyte. Our findings show the importance of understanding processing and the biophysical change in conformation, possibly due to a mechanical or molecular signal, and may aid in the development of a new CSP vaccine.
Collapse
|
50
|
Whitacre DC, Espinosa DA, Peters CJ, Jones JE, Tucker AE, Peterson DL, Zavala FP, Milich DR. P. falciparum and P. vivax Epitope-Focused VLPs Elicit Sterile Immunity to Blood Stage Infections. PLoS One 2015; 10:e0124856. [PMID: 25933001 PMCID: PMC4416889 DOI: 10.1371/journal.pone.0124856] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/17/2015] [Indexed: 01/09/2023] Open
Abstract
In order to design P. falciparum preerythrocytic vaccine candidates, a library of circumsporozoite (CS) T and B cell epitopes displayed on the woodchuck hepatitis virus core antigen (WHcAg) VLP platform was produced. To test the protective efficacy of the WHcAg-CS VLPs, hybrid CS P. berghei/P. falciparum (Pb/Pf) sporozoites were used to challenge immunized mice. VLPs carrying 1 or 2 different CS repeat B cell epitopes and 3 VLPs carrying different CS non-repeat B cell epitopes elicited high levels of anti-insert antibodies (Abs). Whereas, VLPs carrying CS repeat B cell epitopes conferred 98% protection of the liver against a 10,000 Pb/Pf sporozoite challenge, VLPs carrying the CS non-repeat B cell eptiopes were minimally-to-non-protective. One-to-three CS-specific CD4/CD8 T cell sites were also fused to VLPs, which primed CS-specific as well as WHcAg-specific T cells. However, a VLP carrying only the 3 T cell domains failed to protect against a sporozoite challenge, indicating a requirement for anti-CS repeat Abs. A VLP carrying 2 CS repeat B cell epitopes and 3 CS T cell sites in alum adjuvant elicited high titer anti-CS Abs (endpoint dilution titer >1x106) and provided 80–100% protection against blood stage malaria. Using a similar strategy, VLPs were constructed carrying P. vivax CS repeat B cell epitopes (WHc-Pv-78), which elicited high levels of anti-CS Abs and conferred 99% protection of the liver against a 10,000 Pb/Pv sporozoite challenge and elicited sterile immunity to blood stage infection. These results indicate that immunization with epitope-focused VLPs carrying selected B and T cell epitopes from the P. falciparum and P. vivax CS proteins can elicit sterile immunity against blood stage malaria. Hybrid WHcAg-CS VLPs could provide the basis for a bivalent P. falciparum/P. vivax malaria vaccine.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan/immunology
- CD4-Positive T-Lymphocytes/immunology
- Epitopes, B-Lymphocyte/chemistry
- Epitopes, B-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Hepatitis B Virus, Woodchuck/immunology
- Immunity
- Immunization
- Life Cycle Stages
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/prevention & control
- Malaria, Vivax/immunology
- Malaria, Vivax/parasitology
- Malaria, Vivax/prevention & control
- Mice, Inbred C57BL
- Plasmodium falciparum/immunology
- Plasmodium vivax/immunology
- Protozoan Proteins/immunology
- Rabbits
- Repetitive Sequences, Amino Acid
- Reproducibility of Results
- Virion/immunology
Collapse
Affiliation(s)
- David C. Whitacre
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
- VLP Biotech, Inc., San Diego, California, United States of America
| | - Diego A. Espinosa
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Cory J. Peters
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
- VLP Biotech, Inc., San Diego, California, United States of America
| | - Joyce E. Jones
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
- VLP Biotech, Inc., San Diego, California, United States of America
| | - Amy E. Tucker
- VLP Biotech, Inc., San Diego, California, United States of America
| | - Darrell L. Peterson
- Department of Biochemistry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Fidel P. Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - David R. Milich
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
- VLP Biotech, Inc., San Diego, California, United States of America
- * E-mail:
| |
Collapse
|