1
|
Kisalu NK, Silva Pereira LD, Herman JD, Asokan M, Ernste K, Merriam J, Liu C, DeMouth ME, Pegu A, Lofgren M, Dillon M, Bonilla B, MacVicar R, Zur Y, Kiyuka P, Flores-Garcia Y, Chakraborty S, Nikolaeva D, Ogwang R, Flynn B, Francica J, Pierson TC, Koup RA, Zavala F, Wang TT, Alter G, Idris AH, Seder RA. FcγR binding differentially contributes to protection by two human monoclonal antibodies targeting Plasmodium falciparum circumsporozoite protein. Sci Transl Med 2025; 17:eadk6745. [PMID: 40267218 DOI: 10.1126/scitranslmed.adk6745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 09/25/2024] [Accepted: 03/05/2025] [Indexed: 04/25/2025]
Abstract
Antibodies mediate protection against a wide range of pathogens through binding and neutralizing the pathogen or through Fc-mediated effector functions. Human monoclonal antibodies (mAbs) CIS43LS and L9LS show high-affinity binding targeting distinct regions on the Plasmodium falciparum circumsporozoite protein (PfCSP) and are highly effective in preventing malaria in humans. However, the role of FcγR binding in protection by these mAbs has not been determined. Here, we assessed several Fc variants of CIS43LS and L9LS for protection against infection with transgenic Plasmodium berghei parasite expressing PfCSP in mice. Limiting binding to FcγRs did not reduce protection compared to the parental mAbs in mice. To determine whether protection could be improved in vivo by Fc modification, we engineered Fc variant mAbs with increased binding to distinct FcγRs. Passive transfer of CIS43LS-DE and CIS43LS-DEAL variants resulted in an approximately two- to threefold reduction in the liver-stage parasite burden in C57BL/6 or human FcγR mice compared with the parental CIS43LS after challenge. CIS43LS-DEAL also enhanced protection of mice after mosquito bite challenge. Systems serology analysis revealed that the CIS43LS-DE and CIS43LS-DEAL variants could enhance human neutrophil and monocyte phagocytosis, as well as NK cell activation, compared with CIS43LS. However, similar Fc modifications incorporated into L9LS did not increase protection compared to the parental mAb. Overall, although FcγR binding by CIS43LS and L9LS is dispensable in mouse models of malaria, enhancing the binding of CIS43LS to FcγR showed a modest increase in the potency of this mAb.
Collapse
Affiliation(s)
- Neville K Kisalu
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Lais D Silva Pereira
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Jonathan D Herman
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Mangaiarkarasi Asokan
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Keenan Ernste
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Jonah Merriam
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Cuiping Liu
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Megan E DeMouth
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Amarendra Pegu
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Mariah Lofgren
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Marlon Dillon
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian Bonilla
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Ryan MacVicar
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Yonatan Zur
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Patience Kiyuka
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Saborni Chakraborty
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine and Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA
| | - Daria Nikolaeva
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rodney Ogwang
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara Flynn
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Joseph Francica
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Theodore C Pierson
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard A Koup
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Taia T Wang
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine and Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Azza H Idris
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Robert A Seder
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Yoo R, Jore MM, Julien J. Targeting Bottlenecks in Malaria Transmission: Antibody-Epitope Descriptions Guide the Design of Next-Generation Biomedical Interventions. Immunol Rev 2025; 330:e70001. [PMID: 39907429 PMCID: PMC11796336 DOI: 10.1111/imr.70001] [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: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
Malaria continues to pose a significant burden to global health. Thus, a strong need exists for the development of a diverse panel of intervention strategies and modalities to combat malaria and achieve elimination and eradication goals. Deploying interventions that target bottlenecks in the transmission life cycle of the causative agent of malaria, Plasmodium parasites, is an attractive strategy. The development of highly potent antibody-based biologics, including vaccines, can be greatly facilitated by an in-depth molecular understanding of antibody-epitope interactions. Here, we provide an overview of structurally characterized antibodies targeting lead vaccine candidates expressed during the bottlenecks of the Plasmodium life cycle which include the pre-erythrocytic and sexual stages. The repeat region of the circumsporozoite protein (CSP), domain 1 of Pfs230 and domains 1 and 3 of Pfs48/45 are critical Plasmodium regions targeted by the most potent antibodies at the two bottlenecks of transmission, with other promising targets emerging and requiring further characterization.
Collapse
Affiliation(s)
- Randy Yoo
- Program in Molecular MedicineThe Hospital for Sick Children Research InstituteTorontoOntarioCanada
- Department of BiochemistryUniversity of TorontoTorontoOntarioCanada
| | - Matthijs M. Jore
- Department of Medical MicrobiologyRadboudumcNijmegenThe Netherlands
| | - Jean‐Philippe Julien
- Program in Molecular MedicineThe Hospital for Sick Children Research InstituteTorontoOntarioCanada
- Department of BiochemistryUniversity of TorontoTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
3
|
Harris AW, Kurtovic L, Nogueira J, Bouzas I, Opi DH, Wines BD, Lee WS, Hogarth PM, Poumbourios P, Drummer HE, Valim C, Porto LC, Beeson JG. Induction of Fc-dependent functional antibodies against different variants of SARS-CoV-2 varies by vaccine type and prior infection. COMMUNICATIONS MEDICINE 2024; 4:273. [PMID: 39702507 DOI: 10.1038/s43856-024-00686-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 11/21/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND SARS-CoV-2 transmission and COVID-19 disease severity is influenced by immunity from natural infection and/or vaccination. Population-level immunity is complicated by the emergence of viral variants. Antibody Fc-dependent effector functions are as important mediators in immunity. However, their induction in populations with diverse infection and/or vaccination histories and against variants remains poorly defined. METHODS We evaluated Fc-dependent functional antibodies following vaccination with two widely used vaccines, AstraZeneca (AZ) and Sinovac (SV), including antibody binding of Fcγ-receptors and complement-fixation in vaccinated Brazilian adults (n = 222), some of who were previously infected with SARS-CoV-2, as well as adults with natural infection only (n = 200). IgG, IgM, IgA, and IgG subclasses were also quantified. RESULTS AZ induces greater Fcγ-receptor-binding (types I, IIa, and IIIa/b) antibodies than SV or natural infection. Previously infected individuals have significantly greater vaccine-induced responses compared to naïve counterparts. Fcγ-receptor-binding is highest among AZ vaccinated individuals with a prior infection, for all receptor types, and substantial complement-fixing activity is only seen among this group. SV induces higher IgM than AZ, but this does not drive better complement-fixing activity. Some SV responses are associated with subject age, whereas AZ responses are not. Importantly, functional antibody responses are well retained against the Omicron BA.1 S protein, being best retained for Fcγ-receptor-1 binding, and are higher for AZ than SV. CONCLUSIONS Hybrid immunity, from combined natural exposure and vaccination, generates strong Fc-mediated antibody functions which may contribute to immunity against evolving SARS-CoV-2 variants. Understanding determinants of Fc-mediated functions may enable future vaccines with greater efficacy against different variants.
Collapse
Affiliation(s)
- Alexander W Harris
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
| | - Liriye Kurtovic
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
| | - Jeane Nogueira
- Immunogenic and Histocompatibility Laboratory, Technologic Core for Tissue repair and Histocompatibility, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Isabel Bouzas
- Health Research Support Facility Center (CAPCS), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - D Herbert Opi
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
- Departments of Medicine, The University of Melbourne, Melbourne, Australia
| | - Bruce D Wines
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | | | - Pantelis Poumbourios
- Burnet Institute, Melbourne, Australia
- Department of Microbiology, Monash University, Melbourne, Australia
| | - Heidi E Drummer
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Clarissa Valim
- Boston University School of Public Health, Boston University, Boston, USA
| | - Luís Cristóvão Porto
- Immunogenic and Histocompatibility Laboratory, Technologic Core for Tissue repair and Histocompatibility, Rio de Janeiro State University, Rio de Janeiro, Brazil
- Health Research Support Facility Center (CAPCS), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - James G Beeson
- Burnet Institute, Melbourne, Australia.
- Department of Immunology, Monash University, Melbourne, Australia.
- Departments of Medicine, The University of Melbourne, Melbourne, Australia.
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
4
|
Kurtovic L, Feng G, Hysa A, Haghiri A, O'Flaherty K, Wines BD, Santano R, D'Andrea L, Drummer HE, Hogarth PM, Sacarlal J, Fowkes FJI, Simpson JA, Dobaño C, Beeson JG. Antibody mechanisms of protection against malaria in RTS,S-vaccinated children: a post-hoc serological analysis of phase 2 trial. THE LANCET. MICROBE 2024; 5:100898. [PMID: 39127054 DOI: 10.1016/s2666-5247(24)00130-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 03/25/2024] [Accepted: 05/10/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND The RTS,S malaria vaccine is currently recommended for children aged 5-6 months in regions with moderate-to-high Plasmodium falciparum transmission. However, vaccination only confers 55% efficacy over 12 months and wanes within 18 months. The immunological mechanisms of RTS,S-mediated immunity are poorly understood; therefore, we aimed to identify antibody response types associated with protection against malaria in children vaccinated with RTS,S. METHODS In this post-hoc analysis, we evaluated antibody responses in 737 children aged 1-4 years vaccinated with RTS,S in a phase 2b clinical trial conducted in Mozambique in 2003. We evaluated all available samples collected from children 30 days after the three-dose vaccination schedule at study month 3 (M3; n=737 available of 803 children allocated to receive RTS,S). For comparison, we tested a subset of samples collected before vaccination at study month 0 (M0; n=50) and from children in the control vaccine group (M0 n=25; M3 n=99). We quantified the induction of antibodies to different regions of the vaccine antigen that function by fixing serum complement proteins and binding to Fcγ receptors (FcγRs; FcγRI, FcγRIIa, and FcγRIII) expressed on immune cells as potential mechanisms of immunity. FINDINGS Functional antibody responses to the C-terminal region of the vaccine antigen, circumsporozoite protein (CSP), were associated with a reduced risk of malaria (C1q p=0·0060, FcγRIIa p=0·014, and FcγRIII p=0·019). These associations remained significant in male participants when the analyses were stratified by sex (C1q p=0·012, FcγRI p=0·023, FcγRIIa p=0·0070, and FcγRIII p=0·0080). IgA to the central repeat (p=0·0010) and C-terminal (p=0·0040) regions of CSP were also associated with protection. We show that IgA can bind FcαRI and mediate opsonic phagocytosis using a serum pool and monoclonal antibodies. Multiparameter analysis using machine-learning methods suggest that IgA, complement fixation, and FcγRI binding were most predictive of protection against malaria (hazard ratio <1) and suggested that associations differed between male and female participants. INTERPRETATION We provide evidence that functional antibody responses mediated by IgG and IgA are associated with protection against malaria in young children vaccinated with RTS,S, and suggest potential differences in the correlates of immunity between males and females. These findings reveal new avenues that could be used to achieve malaria vaccines with higher efficacy. FUNDING National Health and Medical Research Council, Australia, and Thrasher Research Fund.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Burnet Institute, Melbourne, VIC, Australia; Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Gaoqian Feng
- Burnet Institute, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Alessia Hysa
- Burnet Institute, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Ali Haghiri
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia; School of Engineering, University of Leicester, Leicester, UK
| | | | - Bruce D Wines
- Burnet Institute, Melbourne, VIC, Australia; Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia; Department of Clinical Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Rebeca Santano
- ISGlobal, Hospital Clínic Universitat de Barcelona, Barcelona, Catalonia, Spain; CIBER Enfermedades Infecciosas, Barcelona, Spain
| | | | - Heidi E Drummer
- Burnet Institute, Melbourne, VIC, Australia; Department of Microbiology, Monash University, Melbourne, VIC, Australia; Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - P Mark Hogarth
- Burnet Institute, Melbourne, VIC, Australia; Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia; Department of Clinical Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Jahit Sacarlal
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique; Faculdade de Medicina, Universidade Eduardo Mondlane, Maputo, Mozambique
| | - Freya J I Fowkes
- Burnet Institute, Melbourne, VIC, Australia; Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic Universitat de Barcelona, Barcelona, Catalonia, Spain; CIBER Enfermedades Infecciosas, Barcelona, Spain; Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia; Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia; Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Wang LT, Idris AH, Kisalu NK, Crompton PD, Seder RA. Monoclonal antibodies to the circumsporozoite proteins as an emerging tool for malaria prevention. Nat Immunol 2024; 25:1530-1545. [PMID: 39198635 DOI: 10.1038/s41590-024-01938-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/17/2024] [Indexed: 09/01/2024]
Abstract
Despite various public health strategies, malaria caused by Plasmodium falciparum parasites remains a major global health challenge that requires development of new interventions. Extended half-life human monoclonal antibodies targeting the P. falciparum circumsporozoite protein on sporozoites, the infective form of malaria parasites, prevent malaria in rodents and humans and have been advanced into clinical development. The protective epitopes on the circumsporozoite protein targeted by monoclonal antibodies have been defined. Cryogenic electron and multiphoton microscopy have enabled mechanistic structural and functional investigations of how antibodies bind to the circumsporozoite protein and neutralize sporozoites. Moreover, innovations in bioinformatics and antibody engineering have facilitated enhancement of antibody potency and durability. Here, we summarize the latest scientific advances in understanding how monoclonal antibodies to the circumsporozoite protein prevent malaria and highlight existing clinical data and future plans for how this emerging intervention can be used alone or alongside existing antimalarial interventions to control malaria across at-risk populations.
Collapse
Affiliation(s)
- Lawrence T Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Azza H Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| | - Neville K Kisalu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- PATH's Center for Vaccine Innovation and Access, Washington, DC, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Costa-Gouvea TBL, Françoso KS, Marques RF, Gimenez AM, Faria ACM, Cariste LM, Dominguez MR, Vasconcelos JRC, Nakaya HI, Silveira ELV, Soares IS. Poly I:C elicits broader and stronger humoral and cellular responses to a Plasmodium vivax circumsporozoite protein malaria vaccine than Alhydrogel in mice. Front Immunol 2024; 15:1331474. [PMID: 38650939 PMCID: PMC11033515 DOI: 10.3389/fimmu.2024.1331474] [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: 11/01/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Malaria remains a global health challenge, necessitating the development of effective vaccines. The RTS,S vaccination prevents Plasmodium falciparum (Pf) malaria but is ineffective against Plasmodium vivax (Pv) disease. Herein, we evaluated the murine immunogenicity of a recombinant PvCSP incorporating prevalent polymorphisms, adjuvanted with Alhydrogel or Poly I:C. Both formulations induced prolonged IgG responses, with IgG1 dominance by the Alhydrogel group and high titers of all IgG isotypes by the Poly I:C counterpart. Poly I:C-adjuvanted vaccination increased splenic plasma cells, terminally-differentiated memory cells (MBCs), and precursors relative to the Alhydrogel-combined immunization. Splenic B-cells from Poly I:C-vaccinated mice revealed an antibody-secreting cell- and MBC-differentiating gene expression profile. Biological processes such as antibody folding and secretion were highlighted by the Poly I:C-adjuvanted vaccination. These findings underscore the potential of Poly I:C to strengthen immune responses against Pv malaria.
Collapse
Affiliation(s)
- Tiffany B. L. Costa-Gouvea
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Katia S. Françoso
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rodolfo F. Marques
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alba Marina Gimenez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana C. M. Faria
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Leonardo M. Cariste
- Laboratório de Vacinas Recombinantes, Departamento de Biociências, Universidade Federal de São Paulo, Santos, Brazil
| | - Mariana R. Dominguez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - José Ronnie C. Vasconcelos
- Laboratório de Vacinas Recombinantes, Departamento de Biociências, Universidade Federal de São Paulo, Santos, Brazil
| | - Helder I. Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Institut Pasteur São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Eduardo L. V. Silveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Irene S. Soares
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Dobbs KR, Atieli HE, Valim C, Beeson JG. Previous Malaria Exposures and Immune Dysregulation: Developing Strategies To Improve Malaria Vaccine Efficacy in Young Children. Am J Trop Med Hyg 2024; 110:627-630. [PMID: 38442424 PMCID: PMC10993830 DOI: 10.4269/ajtmh.23-0696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/06/2023] [Indexed: 03/07/2024] Open
Abstract
After several decades in development, two malaria vaccines based on the same antigen and with very similar constructs and adjuvants, RTS,S/AS01 (RTS,S) and R21/Matrix-M (R21), were recommended by the WHO for widespread vaccination of children. These vaccines are much-needed additions to malaria control programs that, when used in conjunction with other control measures, will help to accelerate reductions in malaria morbidity and mortality. Although R21 is not yet available, RTS,S is currently being integrated into routine vaccine schedules in some areas. However, the efficacy of RTS,S is partial, short-lived, and varies widely according to age and geographic location. It is not clear why RTS,S induces protection in some individuals and not others, what the immune mechanisms are that favor protective immunity with RTS,S, and how immune mechanisms are influenced by host and environmental factors. Several studies suggest that higher levels of previous malaria exposure negatively impact RTS,S clinical efficacy. In this article, we summarize data suggesting that previous malaria exposures negatively impact the efficacy of RTS,S and other malaria vaccine candidates. We highlight recent evidence suggesting that increasing malaria exposure impairs the generation of functional antibody responses to RTS,S. Finally, we discuss how investigation of clinical and immune factors associated with suboptimal responses to RTS,S can be used to develop strategies to optimize RTS,S, which will remain relevant to R21 and next-generation vaccines.
Collapse
Affiliation(s)
| | | | - Clarissa Valim
- Boston University School of Public Health, Boston, Massachusetts
| | | |
Collapse
|
8
|
Mutemi DD, Tuju J, Ogwang R, Nyamako L, Wambui KM, Cruz IR, Villner P, Yman V, Kinyanjui SM, Rooth I, Ngasala B, Färnert A, Osier FHA. Antibody-Dependent Respiratory Burst against Plasmodium falciparum Merozoites in Individuals Living in an Area with Declining Malaria Transmission. Vaccines (Basel) 2024; 12:203. [PMID: 38400186 PMCID: PMC10892224 DOI: 10.3390/vaccines12020203] [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: 01/12/2024] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Malaria transmission intensity affects the development of naturally acquired immunity to malaria. An absolute correlate measure of protection against malaria is lacking. However, antibody-mediated functions against Plasmodium falciparum correlate with protection against malaria. In children, antibody-mediated functions against P. falciparum decline with reduced exposure. It is unclear whether adults maintain antibody-mediated functions as malaria transmission declines. This study assessed antibody-dependent respiratory burst (ADRB) in individuals from an area with declining malaria transmission. In an age-matched analysis, we compare ADRB activity during high versus low malaria transmission periods. Age significantly predicted higher ADRB activity in the high (p < 0.001) and low (p < 0.001) malaria transmission periods. ADRB activity was higher during the high compared to the low malaria transmission period in older children and adults. Only older adults during the high malaria transmission period had their median ADRB activity above the ADRB cut-off. Ongoing P. falciparum infection influenced ADRB activity during the low (p = 0.01) but not the high (p = 0.29) malaria transmission period. These findings propose that naturally acquired immunity to P. falciparum is affected in children and adults as malaria transmission declines, implying that vaccines will be necessary to induce and maintain protection against malaria.
Collapse
Affiliation(s)
- Doreen D. Mutemi
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Parasitology and Medical Entomology, Muhimbili University of Health and Allied Sciences, Dar es Salaam 11102, Tanzania
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi 80108, Kenya
| | - James Tuju
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi 80108, Kenya
| | - Rodney Ogwang
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi 80108, Kenya
| | - Lydia Nyamako
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi 80108, Kenya
| | - Kennedy M. Wambui
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi 80108, Kenya
- Epidemiology and Biostatistics Division, School of Public Health, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Ivette R. Cruz
- Division of Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Pär Villner
- Division of Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Victor Yman
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Infectious Diseases, Södersjukhuset, 118 61 Stockholm, Sweden
| | - Samson M. Kinyanjui
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi 80108, Kenya
- Pwani University Bioscience Research Centre, Pwani University, Kilifi 80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LG, UK
- School of Business Studies, Strathmore University, Nairobi 0200, Kenya
| | - Ingegerd Rooth
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, 171 77 Stockholm, Sweden
- Nyamisati Malaria Research Group, Pwani 61621, Tanzania
| | - Billy Ngasala
- Department of Parasitology and Medical Entomology, Muhimbili University of Health and Allied Sciences, Dar es Salaam 11102, Tanzania
- Department of Women’s and Children’s Health, International Maternal and Child Health, Uppsala University, 751 05 Uppsala, Sweden
| | - Anna Färnert
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Faith H. A. Osier
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi 80108, Kenya
- Centre of Infectious Diseases, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
9
|
Mortazavi SE, Lugaajju A, Nylander M, Danielsson L, Tijani MK, Beeson JG, Persson KEM. Acquisition of complement fixing antibodies targeting Plasmodium falciparum merozoites in infants and their mothers in Uganda. Front Immunol 2023; 14:1295543. [PMID: 38090561 PMCID: PMC10715273 DOI: 10.3389/fimmu.2023.1295543] [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: 09/16/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Background Antibody-mediated complement fixation has previously been associated with protection against malaria in naturally acquired immunity. However, the process of early-life development of complement-fixing antibodies in infants, both in comparison to their respective mothers and to other immune parameters, remains less clear. Results We measured complement-fixing antibodies in newborns and their mothers in a malaria endemic area over 5 years follow-up and found that infants' complement-fixing antibody levels were highest at birth, decreased until six months, then increased progressively until they were similar to birth at five years. Infants with high levels at birth experienced a faster decay of complement-fixing antibodies but showed similar levels to the low response group of newborns thereafter. No difference was observed in antibody levels between infant cord blood and mothers at delivery. The same result was found when categorized into high and low response groups, indicating placental transfer of antibodies. Complement-fixing antibodies were positively correlated with total schizont-specific IgG and IgM levels in mothers and infants at several time points. At nine months, complement-fixing antibodies were negatively correlated with total B cell frequency and osteopontin concentrations in the infants, while positively correlated with atypical memory B cells and P. falciparum-positive atypical memory B cells. Conclusion This study indicates that complement-fixing antibodies against P. falciparum merozoites are produced in the mothers and placentally-transferred, and they are acquired in infants over time during the first years of life. Understanding early life immune responses is crucial for developing a functional, long lasting malaria vaccine.
Collapse
Affiliation(s)
- Susanne E. Mortazavi
- Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Infectious Diseases, Skåne University Hospital, Lund, Sweden
| | - Allan Lugaajju
- College of Health Sciences, Makerere University, Kampala, Uganda
| | - Maria Nylander
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Lena Danielsson
- Department of Laboratory Medicine, Lund University, Lund, Sweden
- Clinical Chemistry and Pharmacology, Laboratory Medicine, Office for Medical Services, Region Skåne, Lund, Sweden
| | - Muyideen Kolapo Tijani
- Department of Laboratory Medicine, Lund University, Lund, Sweden
- Cellular Parasitology Program, Cell Biology and Genetics Unit, Department of Zoology, University of Ibadan, Ibadan, Nigeria
| | - James G. Beeson
- The Burnet Institute, Melbourne, VIC, Australia
- Department of Infectious Diseases, University of Melbourne, Melbourne, VIC, Australia
- Central Clinical School and Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - Kristina E. M. Persson
- Department of Laboratory Medicine, Lund University, Lund, Sweden
- Clinical Chemistry and Pharmacology, Laboratory Medicine, Office for Medical Services, Region Skåne, Lund, Sweden
| |
Collapse
|
10
|
Abstract
Malaria is a mosquito-borne disease caused by protozoan parasites of the genus Plasmodium. Despite significant declines in malaria-attributable morbidity and mortality over the last two decades, it remains a major public health burden in many countries. This underscores the critical need for improved strategies to prevent, treat and control malaria if we are to ultimately progress towards the eradication of this disease. Ideally, this will include the development and deployment of a highly effective malaria vaccine that is able to induce long-lasting protective immunity. There are many malaria vaccine candidates in development, with more than a dozen of these in clinical development. RTS,S/AS01 (also known as Mosquirix) is the most advanced malaria vaccine and was shown to have modest efficacy against clinical malaria in phase III trials in 5- to 17-month-old infants. Following pilot implementation trials, the World Health Organisation has recommended it for use in Africa in young children who are most at risk of infection with P. falciparum, the deadliest of the human malaria parasites. It is well recognised that more effective malaria vaccines are needed. In this review, we discuss malaria vaccine candidates that have progressed into clinical evaluation and highlight the most advanced candidates: Sanaria's irradiated sporozoite vaccine (PfSPZ Vaccine), the chemoattenuated sporozoite vaccine (PfSPZ-CVac), RTS,S/AS01 and the novel malaria vaccine candidate, R21, which displayed promising, high-level efficacy in a recent small phase IIb trial in Africa.
Collapse
Affiliation(s)
- Danielle I Stanisic
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| | - Michael F Good
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| |
Collapse
|
11
|
Bell GJ, Gyaase S, Goel V, Adu B, Mensah B, Essone P, Dosoo D, Osei M, Niare K, Wiru K, Brandt K, Emch M, Ghansah A, Asante KP, Mvalo T, Agnandji ST, Juliano JJ, Bailey JA. Background malaria incidence and parasitemia during the three-dose RTS,S/AS01 vaccination series do not reduce magnitude of antibody response nor efficacy against the first case of malaria. BMC Infect Dis 2023; 23:716. [PMID: 37872492 PMCID: PMC10594884 DOI: 10.1186/s12879-023-08699-7] [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: 05/30/2023] [Accepted: 10/11/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND RTS,S/AS01 has been recommended by WHO for widespread implementation in medium to high malaria transmission settings. Previous analyses have noted lower vaccine efficacies in higher transmission settings, possibly due to the more rapid development of naturally acquired immunity in the control group. METHODS To investigate a reduced immune response to vaccination as a potential mechanism behind lower efficacy in high transmission areas, we examine initial vaccine antibody (anti-CSP IgG) response and vaccine efficacy against the first case of malaria (to exclude the effect of naturally acquired immunity) using data from three study areas (Kintampo, Ghana; Lilongwe, Malawi; Lambaréné, Gabon) from the 2009-2014 phase III trial (NCT00866619). Our key exposures are parasitemia during the vaccination series and background malaria incidence. We calculate vaccine efficacy (one minus hazard ratio) using a cox-proportional hazards model and allowing for the time-varying effect of RTS,S/AS01. RESULTS We find that antibody responses to the primary three-dose vaccination series were higher in Ghana than in Malawi and Gabon, but that neither antibody levels nor vaccine efficacy against the first case of malaria varied by background incidence or parasitemia during the primary vaccination series. CONCLUSIONS We find that vaccine efficacy is unrelated to infections during vaccination. Contributing to a conflicting literature, our results suggest that vaccine efficacy is also unrelated to infections before vaccination, meaning that control-group immunity is likely a major reason for lower efficacy in high transmission settings, not reduced immune responses to RTS,S/AS01. This may be reassuring for implementation in high transmission settings, though further studies are needed.
Collapse
Affiliation(s)
- Griffin J Bell
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | | | - Varun Goel
- Carolina Population Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Bright Adu
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Benedicta Mensah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Paulin Essone
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - David Dosoo
- Kintampo Health Research Centre, Kintampo, Ghana
| | - Musah Osei
- Kintampo Health Research Centre, Kintampo, Ghana
| | - Karamoko Niare
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Kenneth Wiru
- Kintampo Health Research Centre, Kintampo, Ghana
| | - Katerina Brandt
- Carolina Population Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Michael Emch
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
- Carolina Population Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Anita Ghansah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | | | - Tisungane Mvalo
- University of North Carolina Project, Lilongwe, Malawi
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Jonathan J Juliano
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
- Division of Infectious Diseases, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Jeffrey A Bailey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| |
Collapse
|
12
|
Rajneesh, Tiwari R, Singh VK, Kumar A, Gupta RP, Singh AK, Gautam V, Kumar R. Advancements and Challenges in Developing Malaria Vaccines: Targeting Multiple Stages of the Parasite Life Cycle. ACS Infect Dis 2023; 9:1795-1814. [PMID: 37708228 DOI: 10.1021/acsinfecdis.3c00332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Malaria, caused by Plasmodium species, remains a major global health concern, causing millions of deaths annually. While the introduction of the RTS,S vaccine has shown promise, there is a pressing need for more effective vaccines due to the emergence of drug-resistant parasites and insecticide-resistant vectors. However, the complex life cycle and genetic diversity of the parasite, technical obstacles, limited funding, and the impact of the 2019 pandemic have hindered progress in malaria vaccine development. This review focuses on advancements in malaria vaccine development, particularly the ongoing clinical trials targeting antigens from different stages of the Plasmodium life cycle. Additionally, we discuss the rationale, strategies, and challenges associated with vaccine design, aiming to enhance the immune response and protective efficacy of vaccine candidates. A cost-effective and multistage vaccine could hold the key to controlling and eradicating malaria.
Collapse
Affiliation(s)
- Rajneesh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rahul Tiwari
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vishal K Singh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Awnish Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rohit P Gupta
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- Department of Applied Microbiology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Akhilesh K Singh
- Faculty of Dental Science, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vibhav Gautam
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rajiv Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
13
|
Rosenkranz M, Fürle K, Hibbert J, Ulmer A, Ali A, Giese T, Blank A, Haefeli WE, Böhnlein E, Lanzer M, Thomson-Luque R. Multifunctional IgG/IgM antibodies and cellular cytotoxicity are elicited by the full-length MSP1 SumayaVac-1 malaria vaccine. NPJ Vaccines 2023; 8:112. [PMID: 37558673 PMCID: PMC10412566 DOI: 10.1038/s41541-023-00701-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 06/27/2023] [Indexed: 08/11/2023] Open
Abstract
Radical control of malaria likely requires a vaccine that targets both the asymptomatic liver stages and the disease-causing blood stages of the human malaria parasite Plasmodium falciparum. While substantial progress has been made towards liver stage vaccines, the development of a blood stage vaccine is lagging behind. We have recently conducted a first-in-human clinical trial to evaluate the safety and immunogenicity of the recombinant, full-length merozoite surface protein 1 (MSP1FL) formulated with GLA-SE as adjuvant. Here, we show that the vaccine, termed SumayaVac-1, elicited both a humoral and cellular immune response as well as a recall T cell memory. The induced IgG and IgM antibodies were able to stimulate various Fc-mediated effector mechanisms associated with protection against malaria, including phagocytosis, release of reactive oxygen species, production of IFN-γ as well as complement activation and fixation. The multifunctional activity of the humoral immune response remained for at least 6 months after vaccination and was comparable to that of naturally acquired anti-MSP1 antibodies from semi-immune adults from Kenya. We further present evidence of SumayaVac-1 eliciting a recallable cellular cytotoxicity by IFN-γ producing CD8+ T cells. Our study revitalizes MSP1FL as a relevant blood stage vaccine candidate and warrants further evaluation of SumayaVac-1 in a phase II efficacy trial.
Collapse
Affiliation(s)
- Micha Rosenkranz
- Center for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kristin Fürle
- Center for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Julia Hibbert
- Center for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anne Ulmer
- Center for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Arin Ali
- Center for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Giese
- Institute for Immunology, Heidelberg University Hospital and German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Antje Blank
- Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Walter E Haefeli
- Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Michael Lanzer
- Center for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Richard Thomson-Luque
- Center for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany.
- Sumaya-Biotech GmbH & Co. KG, Heidelberg, Germany.
| |
Collapse
|
14
|
Vyas VK, Shukla T, Sharma M. Medicinal chemistry approaches for the discovery of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors as antimalarial agents. Future Med Chem 2023; 15:1295-1321. [PMID: 37551689 DOI: 10.4155/fmc-2023-0113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
Malaria is a severe human disease and a global health problem because of drug-resistant strains. Drugs reported to prevent the growth of Plasmodium parasites target various phases of the parasites' life cycle. Antimalarial drugs can inhibit key enzymes that are responsible for the cellular growth and development of parasites. Plasmodium falciparum dihydroorotate dehydrogenase is one such enzyme that is necessary for de novo pyrimidine biosynthesis. This review focuses on various medicinal chemistry approaches used for the discovery and identification of selective P. falciparum dihydroorotate dehydrogenase inhibitors as antimalarial agents. This comprehensive review discusses recent advances in the selective therapeutic activity of distinct chemical classes of compounds as P. falciparum dihydroorotate dehydrogenase inhibitors and antimalarial drugs.
Collapse
Affiliation(s)
- Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Tanvi Shukla
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Manmohan Sharma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| |
Collapse
|
15
|
Ernest M, Rosa TFA, Pala ZR, Kudyba HM, Sweeney B, Reiss T, Pradel G, Vega-Rodríguez J. Plasmodium falciparum Gametes and Sporozoites Hijack Plasmin and Factor H To Evade Host Complement Killing. Microbiol Spectr 2023; 11:e0449322. [PMID: 37191558 PMCID: PMC10269923 DOI: 10.1128/spectrum.04493-22] [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: 11/03/2022] [Accepted: 04/27/2023] [Indexed: 05/17/2023] Open
Abstract
Plasmodium parasites are the etiological agents of malaria, a disease responsible for over half a million deaths annually. Successful completion of the parasite's life cycle in the vertebrate host and transmission to a mosquito vector is contingent upon the ability of the parasite to evade the host's defenses. The extracellular stages of the parasite, including gametes and sporozoites, must evade complement attack in both the mammalian host and in the blood ingested by the mosquito vector. Here, we show that Plasmodium falciparum gametes and sporozoites acquire mammalian plasminogen and activate it into the serine protease plasmin to evade complement attack by degrading C3b. Complement-mediated permeabilization of gametes and sporozoites was higher in plasminogen-depleted plasma, suggesting that plasminogen is important for complement evasion. Plasmin also facilitates gamete exflagellation through complement evasion. Furthermore, supplementing serum with plasmin significantly increased parasite infectivity to mosquitoes and lowered the transmission-blocking activity of antibodies to Pfs230, a potent vaccine candidate currently in clinical trials. Finally, we show that human factor H, previously shown to facilitate complement evasion by gametes, also facilitates complement evasion by sporozoites. Plasmin and factor H simultaneously cooperate to enhance complement evasion by gametes and sporozoites. Taken together, our data show that Plasmodium falciparum gametes and sporozoites hijack the mammalian serine protease plasmin to evade complement attack by degrading C3b. Understanding of the mechanisms of complement evasion by the parasite is key to the development of novel effective therapeutics. IMPORTANCE Current approaches to control malaria are complicated by the development of antimalarial-resistant parasites and insecticide-resistant vectors. Vaccines that block transmission to mosquitoes and humans are a plausible alternative to overcome these setbacks. To inform the development of efficacious vaccines, it is imperative to understand how the parasite interacts with the host immune response. In this report, we show that the parasite can co-opt host plasmin, a mammalian fibrinolytic protein to evade host complement attack. Our results highlight a potential mechanism that may reduce efficacy of potent vaccine candidates. Taken together, our results will inform future studies in developing novel antimalarial therapeutics.
Collapse
Affiliation(s)
- Medard Ernest
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Thiago F. A. Rosa
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Zarna Rajeshkumar Pala
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Heather M. Kudyba
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Brendan Sweeney
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Timo Reiss
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
16
|
Bell GJ, Gyaase S, Goel V, Adu B, Mensah B, Essone P, Dosoo D, Osei M, Niare K, Wiru K, Brandt K, Emch M, Ghansah A, Asante KP, Mvalo T, Agnandji ST, Juliano JJ, Bailey JA. Malaria Transmission Intensity and Parasitemia during the Three-Dose RTS,S/AS01 Vaccination Series do not Reduce Magnitude of Antibody Response nor Efficacy Against the First Case of Malaria. RESEARCH SQUARE 2023:rs.3.rs-2960373. [PMID: 37292711 PMCID: PMC10246269 DOI: 10.21203/rs.3.rs-2960373/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background RTS,S/AS01 has been recommended by WHO for widespread implementation in medium to high malaria transmission settings. Previous analyses have noted lower vaccine efficacies in higher transmission settings, possibly due to the more rapid development of naturally acquired immunity in the control group. Methods To investigate a reduced immune response to vaccination as a potential mechanism behind lower efficacy in high transmission areas, we examine initial vaccine antibody (anti-CSP IgG) response and vaccine efficacy against the first case of malaria to exclude the delayed malaria effect using data from three study areas (Kintampo, Ghana; Lilongwe, Malawi; Lambaréné, Gabon) from the 2009-2014 phase III trial (NCT00866619). Our key exposures are parasitemia during the vaccination series and malaria transmission intensity. We calculate vaccine efficacy (one minus hazard ratio) using a cox-proportional hazards model and allowing for the time-varying effect of RTS,S/AS01. Results We find that antibody responses to the primary three-dose vaccination series were higher in Ghana than in Malawi and Gabon, but that neither antibody levels nor vaccine efficacy against the first case of malaria varied by transmission intensity or parasitemia during the primary vaccination series. Conclusions We find that vaccine efficacy is unrelated to infections during vaccination. Contributing to a conflicting literature, our results suggest that vaccine efficacy is also unrelated to infections before vaccination, meaning that delayed malaria is likely the main reason for lower efficacy in high transmission settings, not reduced immune responses. This may be reassuring for implementation in high transmission settings, though further studies are needed.
Collapse
|
17
|
Defining species-specific and conserved interactions of apical membrane protein 1 during erythrocyte invasion in malaria to inform multi-species vaccines. Cell Mol Life Sci 2023; 80:74. [PMID: 36847896 PMCID: PMC9969379 DOI: 10.1007/s00018-023-04712-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 03/01/2023]
Abstract
Plasmodium falciparum and P. vivax are the major causes of human malaria, and P. knowlesi is an important additional cause in SE Asia. Binding of apical membrane antigen 1 (AMA1) to rhoptry neck protein 2 (RON2) was thought to be essential for merozoite invasion of erythrocytes by Plasmodium spp. Our findings reveal that P. falciparum and P. vivax have diverged and show species-specific binding of AMA1 to RON2, determined by a β-hairpin loop in RON2 and specific residues in AMA1 Loop1E. In contrast, cross-species binding of AMA1 to RON2 is retained between P. vivax and P. knowlesi. Mutation of specific amino acids in AMA1 Loop1E in P. falciparum or P. vivax ablated RON2 binding without impacting erythrocyte invasion. This indicates that the AMA1-RON2-loop interaction is not essential for invasion and additional AMA1 interactions are involved. Mutations in AMA1 that disrupt RON2 binding also enable escape of invasion inhibitory antibodies. Therefore, vaccines and therapeutics will need to be broader than targeting only the AMA1-RON2 interaction. Antibodies targeting AMA1 domain 3 had greater invasion-inhibitory activity when RON2-loop binding was ablated, suggesting this domain is a promising additional target for vaccine development. Targeting multiple AMA1 interactions involved in invasion may enable vaccines that generate more potent inhibitory antibodies and address the capacity for immune evasion. Findings on specific residues for invasion function and species divergence and conservation can inform novel vaccines and therapeutics against malaria caused by three species, including the potential for cross-species vaccines.
Collapse
|
18
|
Ngulube P. Humoral Immune Responses to P. falciparum Circumsporozoite Protein (Pfcsp) Induced by the RTS, S Vaccine - Current Update. Infect Drug Resist 2023; 16:2147-2157. [PMID: 37077252 PMCID: PMC10106824 DOI: 10.2147/idr.s401247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/23/2023] [Indexed: 04/21/2023] Open
Abstract
Malaria vaccines targeting the circumsporozoite protein (CSP) of the P. falciparum parasite have been overall relatively promising. RTS, S is a pre-erythrocytic recombinant protein-based malaria vaccine that targets CSP. RTS, S effectiveness shows some limited success regardless of its 58% efficacy for severe disease. P. falciparum circumsporozoite protein (Pfcsp) has stood to be the main candidate protein for most pre-erythrocytic stage vaccines. Studies on the structural and biophysical characteristics of antibodies specific to CSP (anti-CSP) are underway to achieve fine specificity with the CSP polymorphic regions. More recent studies have proposed the use of different kinds of monoclonal antibodies, the use of appropriate adjuvants, ideal vaccination dose and frequency, and improved targeting of particular epitopes for the robust production of functional antibodies and high complement-fixing activity as other potential methods for achieving long-lasting RTS, S. This review highlights recent findings regarding humoral immune responses to CSP elicited by RTS, S vaccine.
Collapse
Affiliation(s)
- Peter Ngulube
- Department of Biological Sciences, Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
- Correspondence: Peter Ngulube, Email
| |
Collapse
|
19
|
Beeson JG, Kurtovic L, Valim C, Asante KP, Boyle MJ, Mathanga D, Dobano C, Moncunill G. The RTS,S malaria vaccine: Current impact and foundation for the future. Sci Transl Med 2022; 14:eabo6646. [DOI: 10.1126/scitranslmed.abo6646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The RTS,S vaccine has recently been recommended for implementation as a childhood vaccine in regions with moderate-to-high malaria transmission. We discuss mechanisms of vaccine protection and longevity, implementation considerations, and future research needed to increase the vaccine’s health impact, including vaccine modifications for higher efficacy and longevity of protection.
Collapse
Affiliation(s)
- James G. Beeson
- Burnet Institute, Melbourne 3004, Victoria, Australia
- Department of Infectious Diseases, University of Melbourne, Victoria, Australia
- Monash University, Central Clinical School and Department of Microbiology, Victoria, Australia
| | - Liriye Kurtovic
- Burnet Institute, Melbourne 3004, Victoria, Australia
- Monash University, Central Clinical School and Department of Microbiology, Victoria, Australia
| | - Clarissa Valim
- Department of Global Health, Boston University School of Public Health, Boston, MA, USA
| | - Kwaku Poku Asante
- Kintampo Health Research Centre, Kintampo North Municipality, Bono East Region, Ghana
| | - Michelle J. Boyle
- QIMR Berghofer Institute, Herston, Queensland, Australia
- University of Queensland, School of Biomedical Sciences, St Lucia, Queensland, Australia
- Griffith University, Brisbane, Queensland, Australia
| | - Don Mathanga
- Malaria Alert Centre, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Carlota Dobano
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| |
Collapse
|
20
|
Bell GJ, Goel V, Essone P, Dosoo D, Adu B, Mensah BA, Gyaase S, Wiru K, Mougeni F, Osei M, Minsoko P, Sinai C, Niaré K, Juliano JJ, Hudgens M, Ghansah A, Kamthunzi P, Mvalo T, Agnandji ST, Bailey JA, Asante KP, Emch M. Malaria Transmission Intensity Likely Modifies RTS, S/AS01 Efficacy Due to a Rebound Effect in Ghana, Malawi, and Gabon. J Infect Dis 2022; 226:1646-1656. [PMID: 35899811 PMCID: PMC10205900 DOI: 10.1093/infdis/jiac322] [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: 05/20/2022] [Accepted: 07/26/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND RTS,S/AS01 is the first malaria vaccine to be approved and recommended for widespread implementation by the World Health Organization (WHO). Trials reported lower vaccine efficacies in higher-incidence sites, potentially due to a "rebound" in malaria cases in vaccinated children. When naturally acquired protection in the control group rises and vaccine protection in the vaccinated wanes concurrently, malaria incidence can become greater in the vaccinated than in the control group, resulting in negative vaccine efficacies. METHODS Using data from the 2009-2014 phase III trial (NCT00866619) in Lilongwe, Malawi; Kintampo, Ghana; and Lambaréné, Gabon, we evaluate this hypothesis by estimating malaria incidence in each vaccine group over time and in varying transmission settings. After estimating transmission intensities using ecological variables, we fit models with 3-way interactions between vaccination, time, and transmission intensity. RESULTS Over time, incidence decreased in the control group and increased in the vaccine group. Three-dose efficacy in the lowest-transmission-intensity group (0.25 cases per person-year [CPPY]) decreased from 88.2% to 15.0% over 4.5 years, compared with 81.6% to -27.7% in the highest-transmission-intensity group (3 CPPY). CONCLUSIONS These findings suggest that interventions, including the fourth RTS,S dose, that protect vaccinated individuals during the potential rebound period should be implemented for high-transmission settings.
Collapse
Affiliation(s)
- Griffin J Bell
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Varun Goel
- Carolina Population Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Paulin Essone
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - David Dosoo
- Kintampo Health Research Centre, Kintampo, Ghana
| | - Bright Adu
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | | | | | - Kenneth Wiru
- Kintampo Health Research Centre, Kintampo, Ghana
| | - Fabrice Mougeni
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Musah Osei
- Kintampo Health Research Centre, Kintampo, Ghana
| | - Pamela Minsoko
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Cyrus Sinai
- Carolina Population Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Karamoko Niaré
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Jonathan J Juliano
- Division of Infectious Diseases, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Michael Hudgens
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Anita Ghansah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | | | | | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Jeffrey A Bailey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | | | - Michael Emch
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
- Carolina Population Center, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
21
|
Inklaar MR, Barillas-Mury C, Jore MM. Deceiving and escaping complement - the evasive journey of the malaria parasite. Trends Parasitol 2022; 38:962-974. [PMID: 36089499 PMCID: PMC9588674 DOI: 10.1016/j.pt.2022.08.013] [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: 05/13/2022] [Revised: 08/03/2022] [Accepted: 08/19/2022] [Indexed: 01/13/2023]
Abstract
During its life cycle, Plasmodium, the malaria parasite, is exposed to the human and mosquito complement systems. Early experiments demonstrated that activation of complement can pose a serious threat to parasites, but recent studies revealed complement-evasion mechanisms important for parasite survival. Blood-stage parasites and gametes recruit regulators to neutralize human complement activation, while ookinetes inhibit mosquito complement by disrupting epithelial nitration in response to midgut invasion. Here we provide an in-depth overview of the evasion mechanisms currently known and speculate on the existence of others not yet identified. Finally, we discuss how these mechanisms could provide novel targets for urgently needed malaria vaccines and therapeutics.
Collapse
Affiliation(s)
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboudumc, The Netherlands.
| |
Collapse
|
22
|
The impact of human complement on the clinical outcome of malaria infection. Mol Immunol 2022; 151:19-28. [PMID: 36063583 DOI: 10.1016/j.molimm.2022.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 11/21/2022]
Abstract
The tropical disease malaria remains a major cause of global morbidity. Once transmitted to the human by a blood-feeding mosquito, the unicellular malaria parasite comes into contact with the complement system and continues to interact with human complement during its intraerythrocytic replication cycles. In the course of infection, both the classical and the alternative pathway of complement are activated, leading to parasite opsonization and lysis as well as the induction of complement-binding antibodies. While complement activity can be linked to the severity of malaria, it remains to date unclear, whether human complement is beneficial for protective immunity or if extensive complement reactions may rather enhance pathogenesis. In addition, the parasite has evolved molecular strategies to circumvent attack by human complement and has even developed means to utilize complement factors as mediators of host cell infection. In this review, we highlight current knowledge on the role of human complement for the progression of malaria infection. We discuss the various types of interactions between malaria parasites and complement factors with regard to immunity and infection outcome and set a special emphasis on the dual role of complement in the context of parasite fitness.
Collapse
|
23
|
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: 16] [Impact Index Per Article: 5.3] [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
|
24
|
Tursi NJ, Reeder SM, Flores-Garcia Y, Bah MA, Mathis-Torres S, Salgado-Jimenez B, Esquivel R, Xu Z, Chu JD, Humeau L, Patel A, Zavala F, Weiner DB. Engineered DNA-encoded monoclonal antibodies targeting Plasmodium falciparum circumsporozoite protein confer single dose protection in a murine malaria challenge model. Sci Rep 2022; 12:14313. [PMID: 35995959 PMCID: PMC9395511 DOI: 10.1038/s41598-022-18375-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/06/2022] [Indexed: 12/15/2022] Open
Abstract
Novel approaches for malaria prophylaxis remain important. Synthetic DNA-encoded monoclonal antibodies (DMAbs) are a promising approach to generate rapid, direct in vivo host-generated mAbs with potential benefits in production simplicity and distribution coupled with genetic engineering. Here, we explore this approach in a malaria challenge model. We engineered germline-reverted DMAbs based on human mAb clones CIS43, 317, and L9 which target a junctional epitope, major repeat, and minor repeat of the Plasmodium falciparum circumsporozoite protein (CSP) respectively. DMAb variants were encoded into a plasmid vector backbone and their expression and binding profiles were characterized. We demonstrate long-term serological expression of DMAb constructs resulting in in vivo efficacy of CIS43 GL and 317 GL in a rigorous mosquito bite mouse challenge model. Additionally, we engineered an Fc modified variant of CIS43 and L9-based DMAbs to ablate binding to C1q to test the impact of complement-dependent Fc function on challenge outcomes. Complement knockout variant DMAbs demonstrated similar protection to that of WT Fc DMAbs supporting the notion that direct binding to the parasite is sufficient for the protection observed. Further investigation of DMAbs for malaria prophylaxis appears of importance.
Collapse
Affiliation(s)
- Nicholas J Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sophia M Reeder
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mamadou A Bah
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Shamika Mathis-Torres
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Berenice Salgado-Jimenez
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Rianne Esquivel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Ziyang Xu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jacqueline D Chu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Laurent Humeau
- Inovio Pharmaceuticals, Plymouth Meeting, PA, 19462, USA
| | - Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
25
|
Tayipto Y, Rosado J, Gamboa D, White MT, Kiniboro B, Healer J, Opi DH, Beeson JG, Takashima E, Tsuboi T, Harbers M, Robinson L, Mueller I, Longley RJ. Assessment of IgG3 as a serological exposure marker for Plasmodium vivax in areas with moderate-high malaria transmission intensity. Front Cell Infect Microbiol 2022; 12:950909. [PMID: 36017364 PMCID: PMC9395743 DOI: 10.3389/fcimb.2022.950909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Abstract
A more sensitive surveillance tool is needed to identify Plasmodium vivax infections for treatment and to accelerate malaria elimination efforts. To address this challenge, our laboratory has developed an eight-antigen panel that detects total IgG as serological markers of P. vivax exposure within the prior 9 months. The value of these markers has been established for use in areas with low transmission. In moderate-high transmission areas, there is evidence that total IgG is more long-lived than in areas with low transmission, resulting in poorer performance of these markers in these settings. Antibodies that are shorter-lived may be better markers of recent infection for use in moderate-high transmission areas. Using a multiplex assay, the antibody temporal kinetics of total IgG, IgG1, IgG3, and IgM against 29 P. vivax antigens were measured over 36 weeks following asymptomatic P. vivax infection in Papua New Guinean children (n = 31), from an area with moderate-high transmission intensity. IgG3 declined faster to background than total IgG, IgG1, and IgM. Based on these kinetics, IgG3 performance was then assessed for classifying recent exposure in a cohort of Peruvian individuals (n = 590; age 3-85 years) from an area of moderate transmission intensity. Using antibody responses against individual antigens, the highest performance of IgG3 in classifying recent P. vivax infections in the prior 9 months was to one of the Pv-fam-a proteins assessed (PVX_125728) (AUC = 0.764). Surprisingly, total IgG was overall a better marker of recent P. vivax infection, with the highest individual classification performance to RBP2b1986-2653 (PVX_094255) (AUC = 0.838). To understand the acquisition of IgG3 in this Peruvian cohort, relevant epidemiological factors were explored using a regression model. IgG3 levels were positively associated with increasing age, living in an area with (relatively) higher transmission intensity, and having three or more PCR-detected blood-stage P. vivax infections within the prior 13 months. Overall, we found that IgG3 did not have high accuracy for detecting recent exposure to P. vivax in the Peruvian cohort, with our data suggesting that this is due to the high levels of prior exposure required to acquire high IgG3 antibody levels.
Collapse
Affiliation(s)
- Yanie Tayipto
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Jason Rosado
- Unité Malaria: Parasites et Hôtes, Département Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France
| | - Dionicia Gamboa
- Laboratorio International Centers of Excellence for Malaria Research (ICEMR)-Amazonia, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Michael T. White
- Unité Malaria: Parasites et Hôtes, Département Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France
| | - Benson Kiniboro
- Vector Borne Disease Unit, Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Julie Healer
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - D. Herbert Opi
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
- Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - James G. Beeson
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
- Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, VIC, Australia
- Department of Microbiology and Central Clinical School, Monash University, Clayton, VIC, Australia
| | - Eizo Takashima
- Proteo-Science Center, Ehime University, Matsuyama, Japan
| | | | - Matthias Harbers
- CellFree Sciences Co., Ltd., Yokohama, Japan
- RIKEN Centre for Integrative Medical Sciences, Yokohama, Japan
| | - Leanne Robinson
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
| | - Ivo Mueller
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Rhea J. Longley
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
26
|
Wines BD, Kurtovic L, Trist HM, Esparon S, Lopez E, Chappin K, Chan LJ, Mordant FL, Lee WS, Gherardin NA, Patel SK, Hartley GE, Pymm P, Cooney JP, Beeson JG, Godfrey DI, Burrell LM, van Zelm MC, Wheatley AK, Chung AW, Tham WH, Subbarao K, Kent SJ, Hogarth PM. Fc engineered ACE2-Fc is a potent multifunctional agent targeting SARS-CoV2. Front Immunol 2022; 13:889372. [PMID: 35967361 PMCID: PMC9369017 DOI: 10.3389/fimmu.2022.889372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/27/2022] [Indexed: 01/26/2023] Open
Abstract
Joining a function-enhanced Fc-portion of human IgG to the SARS-CoV-2 entry receptor ACE2 produces an antiviral decoy with strain transcending virus neutralizing activity. SARS-CoV-2 neutralization and Fc-effector functions of ACE2-Fc decoy proteins, formatted with or without the ACE2 collectrin domain, were optimized by Fc-modification. The different Fc-modifications resulted in distinct effects on neutralization and effector functions. H429Y, a point mutation outside the binding sites for FcγRs or complement caused non-covalent oligomerization of the ACE2-Fc decoy proteins, abrogated FcγR interaction and enhanced SARS-CoV-2 neutralization. Another Fc mutation, H429F did not improve virus neutralization but resulted in increased C5b-C9 fixation and transformed ACE2-Fc to a potent mediator of complement-dependent cytotoxicity (CDC) against SARS-CoV-2 spike (S) expressing cells. Furthermore, modification of the Fc-glycan enhanced cell activation via FcγRIIIa. These different immune profiles demonstrate the capacity of Fc-based agents to be engineered to optimize different mechanisms of protection for SARS-CoV-2 and potentially other viral pathogens.
Collapse
Affiliation(s)
- Bruce D. Wines
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Liriye Kurtovic
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Halina M. Trist
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
| | - Sandra Esparon
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
| | - Ester Lopez
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Klasina Chappin
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
| | - Li-Jin Chan
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Francesca L. Mordant
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Nicholas A. Gherardin
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Sheila K. Patel
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Gemma E. Hartley
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Phillip Pymm
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - James P. Cooney
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - James G. Beeson
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, 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
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Louise M. Burrell
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Menno C. van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Alfred Hospital, Melbourne, VIC, Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Australian Research Council Centre for Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Melbourne, VIC, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Wai-Hong Tham
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- World Health Organization (WHO) Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Australian Research Council Centre for Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - P. Mark Hogarth
- Immune therapies Laboratory, Burnet Institute, Melbourne, VIC, Australia
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
27
|
Chan JA, Loughland JR, de la Parte L, Okano S, Ssewanyana I, Nalubega M, Nankya F, Musinguzi K, Rek J, Arinaitwe E, Tipping P, Bourke P, Andrew D, Dooley N, SheelaNair A, Wines BD, Hogarth PM, Beeson JG, Greenhouse B, Dorsey G, Kamya M, Hartel G, Minigo G, Feeney M, Jagannathan P, Boyle MJ. Age-dependent changes in circulating Tfh cells influence development of functional malaria antibodies in children. Nat Commun 2022; 13:4159. [PMID: 35851033 PMCID: PMC9293980 DOI: 10.1038/s41467-022-31880-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 07/08/2022] [Indexed: 01/29/2023] Open
Abstract
T-follicular helper (Tfh) cells are key drivers of antibodies that protect from malaria. However, little is known regarding the host and parasite factors that influence Tfh and functional antibody development. Here, we use samples from a large cross-sectional study of children residing in an area of high malaria transmission in Uganda to characterize Tfh cells and functional antibodies to multiple parasites stages. We identify a dramatic re-distribution of the Tfh cell compartment with age that is independent of malaria exposure, with Th2-Tfh cells predominating in early childhood, while Th1-Tfh cell gradually increase to adult levels over the first decade of life. Functional antibody acquisition is age-dependent and hierarchical acquired based on parasite stage, with merozoite responses followed by sporozoite and gametocyte antibodies. Antibodies are boosted in children with current infection, and are higher in females. The children with the very highest antibody levels have increased Tfh cell activation and proliferation, consistent with a key role of Tfh cells in antibody development. Together, these data reveal a complex relationship between the circulating Tfh compartment, antibody development and protection from malaria.
Collapse
Affiliation(s)
- Jo-Anne Chan
- Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Jessica R Loughland
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia
| | | | - Satomi Okano
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Isaac Ssewanyana
- Infectious Diseases Research Collaboration, Kampala, Uganda
- London School of Hygiene and Tropical Medicine, London, UK
| | - Mayimuna Nalubega
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | | | | | - John Rek
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Peta Tipping
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia
| | - Peter Bourke
- Division of Medicine, Cairns Hospital, Manunda, QLD, Australia
| | - Dean Andrew
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Nicholas Dooley
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
- Griffith University, Brisbane, QLD, Australia
| | - Arya SheelaNair
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Bruce D Wines
- Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - P Mark Hogarth
- Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
- Department of Microbiology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | | | - Grant Dorsey
- University of California San Francisco, San Francisco, CA, USA
| | - Moses Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Gunter Hartel
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Gabriela Minigo
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia
- College of Health and Human Sciences, Charles Darwin University, Darwin, NT, Australia
| | - Margaret Feeney
- University of California San Francisco, San Francisco, CA, USA
| | | | - Michelle J Boyle
- Burnet Institute, Melbourne, VIC, Australia.
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia.
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
- Griffith University, Brisbane, QLD, Australia.
| |
Collapse
|
28
|
Mugo RM, Mwai K, Mwacharo J, Shee FM, Musyoki JN, Wambua J, Otieno E, Bejon P, Ndungu FM. Seven-year kinetics of RTS, S/AS01-induced anti-CSP antibodies in young Kenyan children. Malar J 2021; 20:452. [PMID: 34856981 PMCID: PMC8641151 DOI: 10.1186/s12936-021-03961-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/20/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND RTS,S/AS01, the leading malaria vaccine has been recommended by the WHO for widespread immunization of children at risk. RTS,S/AS01-induced anti-CSP IgG antibodies are associated with the vaccine efficacy. Here, the long-term kinetics of RTS,S/AS01-induced antibodies was investigated. METHODS 150 participants were randomly selected from the 447 children who participated in the RTS,S/AS01 phase IIb clinical trial in 2007 from Kilifi-Kenya. Cumulatively, the retrospective follow-up period was 93 months with annual plasma samples collection. The levels of anti-CSP IgM, total IgG, IgG1, IgG2, IgG3, and IgG4 antibodies were then determined using an enzyme-linked immunosorbent assay. RESULTS RTS,S/AS01 induced high levels of anti-CSP IgG antibodies which exhibited a rapid waning over 6.5 months post-vaccination, followed by a slower decay over the subsequent years. RTS,S/AS01-induced anti-CSP IgG antibodies remained elevated above the control group levels throughout the 7 years follow-up period. The anti-CSP IgG antibodies were mostly IgG1, IgG3, IgG2, and to a lesser extent IgG4. IgG2 predominated in later timepoints. RTS,S/AS01 also induced high levels of anti-CSP IgM antibodies which increased above the control group levels by month 3. The controls exhibited increasing levels of the anti-CSP IgM antibodies which caught up with the RTS,S/AS01 vaccinees levels by month 21. In contrast, there were no measurable anti-CSP IgG antibodies among the controls. CONCLUSION RTS,S/AS01-induced anti-CSP IgG antibodies kinetics are consistent with long-lived but waning vaccine efficacy. Natural exposure induces anti-CSP IgM antibodies in children, which increases with age, but does not induce substantial levels of anti-CSP IgG antibodies.
Collapse
Affiliation(s)
- Robert M Mugo
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya.
- Institute of Immunology, Center for Infection Medicine, Freie Universtät Berlin, 14163, Berlin, Germany.
- Department of Biological Sciences, Pwani University, P.O. Box 195-80108, Kilifi, Kenya.
| | - Kennedy Mwai
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Jedidah Mwacharo
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Faiz M Shee
- Department of Biological Sciences, Pwani University, P.O. Box 195-80108, Kilifi, Kenya
| | - Jennifer N Musyoki
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Juliana Wambua
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Edward Otieno
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
| | - Philip Bejon
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Francis M Ndungu
- KEMRI-Wellcome Trust Collaborative Programme, P.O. Box 230, Kilifi, 80108, Kenya.
- Institute of Immunology, Center for Infection Medicine, Freie Universtät Berlin, 14163, Berlin, Germany.
- Department of Biological Sciences, Pwani University, P.O. Box 195-80108, Kilifi, Kenya.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Division of Infectious Diseases, Department of Medicine Solna and Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
29
|
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: 3.3] [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
|
30
|
Wang LT, Pereira LS, Kiyuka PK, Schön A, Kisalu NK, Vistein R, Dillon M, Bonilla BG, Molina-Cruz A, Barillas-Mury C, Tan J, Idris AH, Francica JR, Seder RA. Protective effects of combining monoclonal antibodies and vaccines against the Plasmodium falciparum circumsporozoite protein. PLoS Pathog 2021; 17:e1010133. [PMID: 34871332 PMCID: PMC8675929 DOI: 10.1371/journal.ppat.1010133] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/16/2021] [Accepted: 11/19/2021] [Indexed: 11/18/2022] Open
Abstract
Combinations of monoclonal antibodies (mAbs) against different epitopes on the same antigen synergistically neutralize many viruses. However, there are limited studies assessing whether combining human mAbs against distinct regions of the Plasmodium falciparum (Pf) circumsporozoite protein (CSP) enhances in vivo protection against malaria compared to each mAb alone or whether passive transfer of PfCSP mAbs would improve protection following vaccination against PfCSP. Here, we isolated a panel of human mAbs against the subdominant C-terminal domain of PfCSP (C-CSP) from a volunteer immunized with radiation-attenuated Pf sporozoites. These C-CSP-specific mAbs had limited binding to sporozoites in vitro that was increased by combination with neutralizing human "repeat" mAbs against the NPDP/NVDP/NANP tetrapeptides in the central repeat region of PfCSP. Nevertheless, passive transfer of repeat- and C-CSP-specific mAb combinations did not provide enhanced protection against in vivo sporozoite challenge compared to repeat mAbs alone. Furthermore, combining potent repeat-specific mAbs (CIS43, L9, and 317) that respectively target the three tetrapeptides (NPDP/NVDP/NANP) did not provide additional protection against in vivo sporozoite challenge. However, administration of either CIS43, L9, or 317 (but not C-CSP-specific mAbs) to mice that had been immunized with R21, a PfCSP-based virus-like particle vaccine that induces polyclonal antibodies against the repeat region and C-CSP, provided enhanced protection against sporozoite challenge when compared to vaccine or mAbs alone. Collectively, this study shows that while combining mAbs against the repeat and C-terminal regions of PfCSP provide no additional protection in vivo, repeat mAbs do provide increased protection when combined with vaccine-induced polyclonal antibodies. These data should inform the implementation of PfCSP human mAbs alone or following vaccination to prevent malaria infection.
Collapse
Affiliation(s)
- Lawrence T. Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lais S. Pereira
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Patience K. Kiyuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Biological Sciences, Pwani University, Kilifi, Kenya
| | - Arne Schön
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Neville K. Kisalu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rachel Vistein
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Marlon Dillon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Brian G. Bonilla
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alvaro Molina-Cruz
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Joshua Tan
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Azza H. Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
| | - Joseph R. Francica
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert A. Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
31
|
Das J, Fallon JK, Yu TC, Michell A, Suscovich TJ, Linde C, Natarajan H, Weiner J, Coccia M, Gregory S, Ackerman ME, Bergmann-Leitner E, Fontana L, Dutta S, Lauffenburger DA, Jongert E, Wille-Reece U, Alter G. Delayed fractional dosing with RTS,S/AS01 improves humoral immunity to malaria via a balance of polyfunctional NANP6- and Pf16-specific antibodies. MED 2021; 2:1269-1286.e9. [DOI: 10.1016/j.medj.2021.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 07/01/2021] [Accepted: 10/07/2021] [Indexed: 02/06/2023]
|
32
|
Tayipto Y, Liu Z, Mueller I, Longley RJ. Serology for Plasmodium vivax surveillance: A novel approach to accelerate towards elimination. Parasitol Int 2021; 87:102492. [PMID: 34728377 DOI: 10.1016/j.parint.2021.102492] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/30/2021] [Accepted: 10/28/2021] [Indexed: 01/13/2023]
Abstract
Plasmodium vivax is the most widespread causative agent of human malaria in the world. Despite the ongoing implementation of malaria control programs, the rate of case reduction has declined over the last 5 years. Hence, surveillance of malaria transmission should be in place to identify and monitor areas that require intensified malaria control interventions. Serological tools may offer additional insights into transmission intensity over parasite and entomological measures, especially as transmission levels decline. Antibodies can be detected in the host system for months to even years after parasite infections have been cleared from the blood, enabling malaria exposure history to be captured. Because the Plasmodium parasite expresses more than 5000 proteins, it is important to a) understand antibody longevity following infection and b) measure antibodies to more than one antigen in order to accurately inform on the exposure and/or immune status of populations. This review summarises current practices for surveillance of P. vivax malaria, the current state of research into serological exposure markers and their potential role for accelerating malaria elimination, and discusses further studies that need to be undertaken to see such technology implemented in malaria-endemic areas.
Collapse
Affiliation(s)
- Yanie Tayipto
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Zoe Liu
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong, Victoria, Australia; School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong, Australia
| | - Ivo Mueller
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Rhea J Longley
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
33
|
Identification of first-in-class plasmodium OTU inhibitors with potent anti-malarial activity. Biochem J 2021; 478:3445-3466. [PMID: 34486667 DOI: 10.1042/bcj20210481] [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: 06/23/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 11/17/2022]
Abstract
OTU proteases antagonize the cellular defense in the host cells and involve in pathogenesis. Intriguingly, P. falciparum, P. vivax, and P. yoelii have an uncharacterized and highly conserved viral OTU-like proteins. However, their structure, function or inhibitors have not been previously reported. To this end, we have performed structural modeling, small molecule screening, deconjugation assays to characterize and develop first-in-class inhibitors of P. falciparum, P. vivax, and P. yoelii OTU-like proteins. These Plasmodium OTU-like proteins have highly conserved residues in the catalytic and inhibition pockets similar to viral OTU proteins. Plasmodium OTU proteins demonstrated Ubiquitin and ISG15 deconjugation activities as evident by intracellular ubiquitinated protein content analyzed by western blot and flow cytometry. We screened a library of small molecules to determine plasmodium OTU inhibitors with potent anti-malarial activity. Enrichment and correlation studies identified structurally similar molecules. We have identified two small molecules that inhibit P. falciparum, P. vivax, and P. yoelii OTU proteins (IC50 values as low as 30 nM) with potent anti-malarial activity (IC50 of 4.1-6.5 µM). We also established enzyme kinetics, druglikeness, ADME, and QSAR model. MD simulations allowed us to resolve how inhibitors interacted with plasmodium OTU proteins. These findings suggest that targeting malarial OTU-like proteases is a plausible strategy to develop new anti-malarial therapies.
Collapse
|
34
|
Opi DH, Kurtovic L, Chan JA, Horton JL, Feng G, Beeson JG. Multi-functional antibody profiling for malaria vaccine development and evaluation. Expert Rev Vaccines 2021; 20:1257-1272. [PMID: 34530671 DOI: 10.1080/14760584.2021.1981864] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION A vaccine would greatly accelerate current global efforts toward malaria elimination. While a partially efficacious vaccine has been achieved for Plasmodium falciparum, a major bottleneck in developing highly efficacious vaccines is a lack of reliable correlates of protection, and the limited application of assays that quantify functional immune responses to evaluate and down-select vaccine candidates in pre-clinical studies and clinical trials. AREAS COVERED In this review, we describe the important role of antibodies in immunity against malaria and detail the nature and functional activities of antibodies against the malaria-causing parasite. We highlight the growing understanding of antibody effector functions against malaria and in vitro assays to measure these functional antibody responses. We discuss the application of these assays to quantify antibody functions in vaccine development and evaluation. EXPERT OPINION It is becoming increasingly clear that multiple antibody effector functions are involved in immunity to malaria. Therefore, we propose that evaluating vaccine candidates needs to move beyond individual assays or measuring IgG magnitude alone. Instead, vaccine evaluation should incorporate the full breadth of antibody response types and harness a wider range of assays measuring functional antibody responses. We propose a 3-tier approach to implementing assays to inform vaccine evaluation.
Collapse
Affiliation(s)
- D Herbert Opi
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Liriye Kurtovic
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Jo-Anne Chan
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Jessica L Horton
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Gaoqian Feng
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - James G Beeson
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia.,Department of Microbiology, Monash University, Clayton, Australia
| |
Collapse
|
35
|
Asghari A, Nourmohammadi H, Majidiani H, Shariatzadeh SA, Shams M, Montazeri F. In silico analysis and prediction of immunogenic epitopes for pre-erythrocytic proteins of the deadly Plasmodium falciparum. INFECTION GENETICS AND EVOLUTION 2021; 93:104985. [PMID: 34214673 DOI: 10.1016/j.meegid.2021.104985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/23/2021] [Accepted: 06/27/2021] [Indexed: 12/18/2022]
Abstract
Malaria is the deadliest parasitic disease in tropical and subtropical areas around the world, with considerable morbidity and mortality, particularly due to the life-threatening Plasmodium falciparum. The present in silico investigation was performed to reveal the biophysical characteristics and immunogenic epitopes of the six pre-erythrocytic proteins of the P. falciparum using comprehensive immunoinformatics approaches. For this aim, different web servers were employed to predict subcellular localization, antigenicity, allergenicity, solubility, physico-chemical properties, post-translational modification sites (PTMs), the presence of signal peptide and transmembrane domains. Moreover, the secondary and tertiary structures of the proteins were revealed followed by refinement and validations. Finally, NetCTL server was used to predict cytotoxic T-lymphocyte (CTL) epitopes, followed by subsequent screening in terms of antigenicity and immunogenicity. Also, IEDB server was utilized to predict helper T-lymphocyte (HTL) epitopes, followed by screening regarding interferon gamma induction and population coverage. These proteins showed appropriate antigenicity, abundant PTMs as well as many CTL and HTL epitopes, which could be directed for future vaccination studies in the context of multi-epitope vaccine design.
Collapse
Affiliation(s)
- Ali Asghari
- Department of Medical Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Nourmohammadi
- Department of Internal Medicine, Shahid Mostafa Khomeini Hospital, Ilam University of Medical Sciences, Ilam, Iran; Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Hamidreza Majidiani
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Seyyed Ali Shariatzadeh
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Toxoplasmosis Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Morteza Shams
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran; Student Research Committee, Ilam University of Medical Sciences, Ilam, Iran.
| | - Fattaneh Montazeri
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
36
|
Rathnayake D, Aitken EH, Rogerson SJ. Beyond Binding: The Outcomes of Antibody-Dependent Complement Activation in Human Malaria. Front Immunol 2021; 12:683404. [PMID: 34168652 PMCID: PMC8217965 DOI: 10.3389/fimmu.2021.683404] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 05/24/2021] [Indexed: 11/13/2022] Open
Abstract
Antibody immunity against malaria is effective but non-sterile. In addition to antibody-mediated inhibition, neutralisation or opsonisation of malaria parasites, antibody-mediated complement activation is also important in defense against infection. Antibodies form immune complexes with parasite-derived antigens that can activate the classical complement pathway. The complement system provides efficient surveillance for infection, and its activation leads to parasite lysis or parasite opsonisation for phagocytosis. The induction of complement-fixing antibodies contributes significantly to the development of protective immunity against clinical malaria. These complement-fixing antibodies can form immune complexes that are recognised by complement receptors on innate cells of the immune system. The efficient clearance of immune complexes is accompanied by complement receptor internalisation, abrogating the detrimental consequences of excess complement activation. Here, we review the mechanisms of activation of complement by alternative, classical, and lectin pathways in human malaria at different stages of the Plasmodium life cycle with special emphasis on how complement-fixing antibodies contribute to protective immunity. We briefly touch upon the action of anaphylatoxins, the assembly of membrane attack complex, and the possible reasons underlying the resistance of infected erythrocytes towards antibody-mediated complement lysis, relevant to their prolonged survival in the blood of the human host. We make suggestions for further research on effector functions of antibody-mediated complement activation that would guide future researchers in deploying complement-fixing antibodies in preventive or therapeutic strategies against malaria.
Collapse
Affiliation(s)
| | | | - Stephen J. Rogerson
- Department of Infectious Diseases, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
37
|
Napodano C, Marino M, Stefanile A, Pocino K, Scatena R, Gulli F, Rapaccini GL, Delli Noci S, Capozio G, Rigante D, Basile U. Immunological Role of IgG Subclasses. Immunol Invest 2021; 50:427-444. [PMID: 32522062 DOI: 10.1080/08820139.2020.1775643] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The loss of tolerance to self-antigens is the unequivocal "red line" of autoimmunity: both development of autoreactive T and B cells and production of polyclonal autoantibodies represent seminal keys to the pathogenesis of protean autoimmune diseases. Most of these autoantibodies are immunoglobulins G (IgG), functionally distinguished in four subclasses named IgG1, IgG2, IgG3, and IgG4, due to structural differences in the hinge and heavy chain constant regions. Different studies analyzed serum levels of IgG subclasses in the course of different disorders, showing that they might have a pathogenic role by regulating interactions among immunoglobulins, Fc-gamma receptors, and complement. To date, the mechanisms promoting different IgG subclasses distribution during the natural history of most autoimmune diseases remain somewhat unclear. Evidence from the medical literature shows that the serum IgG profile is peculiar for many autoimmune diseases, suggesting that different subclasses could be specific for the underlying driving autoantigens. A better knowledge of IgG subsets may probably help to elucidate their pathological task, but also to define their relevance for diagnostic purposes, patients' personalized management, and prognosis assessment.
Collapse
Affiliation(s)
- Cecilia Napodano
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - MariaPaola Marino
- Institute of General Pathology, Dipartimento Di Medicina E Chirurgia Traslazionale, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Annunziata Stefanile
- Area Diagnostica di Laboratorio, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Krizia Pocino
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Roberto Scatena
- Dipartimento Di Medicina Di Laboratorio, Ospedale Madre Giuseppina Vannini, Rome, Italy
| | - Francesca Gulli
- Dipartimento Di Medicina Di Laboratorio, Ospedale Madre Giuseppina Vannini, Rome, Italy
| | - Gian Lodovico Rapaccini
- Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Delli Noci
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Giovanna Capozio
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Donato Rigante
- Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Umberto Basile
- Area Diagnostica di Laboratorio, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
38
|
Maharaj L, Adeleke VT, Fatoba AJ, Adeniyi AA, Tshilwane SI, Adeleke MA, Maharaj R, Okpeku M. Immunoinformatics approach for multi-epitope vaccine design against P. falciparum malaria. INFECTION GENETICS AND EVOLUTION 2021; 92:104875. [PMID: 33905890 DOI: 10.1016/j.meegid.2021.104875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 04/07/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022]
Abstract
Plasmodium falciparum (P. falciparum) is a leading causative agent of malaria, an infectious disease that can be fatal. Unfortunately, control measures are becoming less effective over time. A vaccine is needed to effectively control malaria and lead towards the total elimination of the disease. There have been multiple attempts to develop a vaccine, but to date, none have been certified as appropriate for wide-scale use. In this study, an immunoinformatics method is presented to design a multi-epitope vaccine construct predicted to be effective against P. falciparum malaria. This was done through the prediction of 12 CD4+ T-cell, 10 CD8+ T-cell epitopes and, 1 B-cell epitope which were assessed for predicted high antigenicity, immunogenicity, and non-allergenicity through in silico methods. The Human Leukocyte Antigen (HLA) population coverage showed that the alleles associated with the epitopes accounted for 78.48% of the global population. The CD4+ and CD8+ T-cell epitopes were docked to HLA-DRB1*07:01 and HLA-A*32:01 successfully. Therefore, the epitopes were deemed to be suitable as components of a multi-epitope vaccine construct. Adjuvant RS09 was added to the construct to generate a stronger immune response, as confirmed by an immune system simulation. Finally, the structural stability of the predicted multi-epitope vaccine was assessed using molecular dynamics simulations. The results show a promising vaccine design that should be further synthesised and assessed for its efficacy in an experimental laboratory setting.
Collapse
Affiliation(s)
- Leah Maharaj
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Victoria T Adeleke
- Discipline of Chemical Engineering, University of KwaZulu-Natal, Howard Campus, Durban 4041, South Africa
| | - Abiodun J Fatoba
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Adebayo A Adeniyi
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, South Africa; Department of Industrial Chemistry, Federal University Oye Ekiti, Nigeria
| | - Selaelo I Tshilwane
- School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Matthew A Adeleke
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Rajendra Maharaj
- Office of Malaria Research, Medical Research Council, South Africa
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa.
| |
Collapse
|
39
|
Soon MSF, Nalubega M, Boyle MJ. T-follicular helper cells in malaria infection and roles in antibody induction. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab008. [PMID: 36845571 PMCID: PMC9914587 DOI: 10.1093/oxfimm/iqab008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 01/29/2023] Open
Abstract
Immunity to malaria is mediated by antibodies that block parasite replication to limit parasite burden and prevent disease. Cytophilic antibodies have been consistently shown to be associated with protection, and recent work has improved our understanding of the direct and Fc-mediated mechanisms of protective antibodies. Antibodies also have important roles in vaccine-mediated immunity. Antibody induction is driven by the specialized CD4+ T cells, T-follicular helper (Tfh) cells, which function within the germinal centre to drive B-cell activation and antibody induction. In humans, circulating Tfh cells can be identified in peripheral blood and are differentiated into subsets that appear to have pathogen/vaccination-specific roles in antibody induction. Tfh cell responses are essential for protective immunity from Plasmodium infection in murine models of malaria. Our understanding of the activation of Tfh cells during human malaria infection and the importance of different Tfh cell subsets in antibody development is still emerging. This review will discuss our current knowledge of Tfh cell activation and development in malaria, and the potential avenues and pitfalls of targeting Tfh cells to improve malaria vaccines.
Collapse
Affiliation(s)
- Megan S F Soon
- Department of Infectious Diseases, QIMR-Berghofer, 300 Herston Road, Herston, QLD, 4006, Australia
| | - Mayimuna Nalubega
- Infectious Diseases Research Collaboration, Tororo District Hospital, Tororo, Uganda
| | - Michelle J Boyle
- Department of Infectious Diseases, QIMR-Berghofer, 300 Herston Road, Herston, QLD, 4006, Australia,Correspondence address. QIMR Berghofer Medical Research Institute, Brisbane, Australia. E-mail:
| |
Collapse
|
40
|
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: 10] [Impact Index Per Article: 2.5] [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
|
41
|
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: 2.0] [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
|
42
|
Abstract
Introduction: An effective vaccine against malaria forms a global health priority. Both naturally acquired immunity and sterile protection induced by irradiated sporozoite immunization were described decades ago. Still no vaccine exists that sufficiently protects children in endemic areas. Identifying immunological correlates of vaccine efficacy can inform rational vaccine design and potentially accelerate clinical development.Areas covered: We discuss recent research on immunological correlates of malaria vaccine efficacy, including: insights from state-of-the-art omics platforms and systems vaccinology analyses; functional anti-parasitic assays; pre-immunization predictors of vaccine efficacy; and comparison of correlates of vaccine efficacy against controlled human malaria infections (CHMI) and against naturally acquired infections.Expert Opinion: Effective vaccination may be achievable without necessarily understanding immunological correlates, but the relatively disappointing efficacy of malaria vaccine candidates in target populations is concerning. Hypothesis-generating omics and systems vaccinology analyses, alongside assessment of pre-immunization correlates, have the potential to bring about paradigm-shifts in malaria vaccinology. Functional assays may represent in vivo effector mechanisms, but have scarcely been formally assessed as correlates. Crucially, evidence is still meager that correlates of vaccine efficacy against CHMI correspond with those against naturally acquired infections in target populations. Finally, the diversity of immunological assays and efficacy endpoints across malaria vaccine trials remains a major confounder.
Collapse
Affiliation(s)
| | - Matthew B B McCall
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
43
|
Ssewanyana I, Rek J, Rodriguez I, Wu L, Arinaitwe E, Nankabirwa JI, Beeson JG, Mayanja-Kizza H, Rosenthal PJ, Dorsey G, Kamya MR, Drakeley C, Greenhouse B, Tetteh KKA. Impact of a Rapid Decline in Malaria Transmission on Antimalarial IgG Subclasses and Avidity. Front Immunol 2021; 11:576663. [PMID: 33584643 PMCID: PMC7873448 DOI: 10.3389/fimmu.2020.576663] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/17/2020] [Indexed: 11/19/2022] Open
Abstract
Understanding how immunity to malaria is affected by declining transmission is important to aid vaccine design and understand disease resurgence. Both IgG subclasses and avidity of antigen-specific responses are important components of an effective immune response. Using a multiplex bead array assay, we measured the total IgG, IgG subclasses, and avidity profiles of responses to 18 P. falciparum blood stage antigens in samples from 160 Ugandans collected at two time points during high malaria transmission and two time points following a dramatic reduction in transmission. Results demonstrated that, for the antigens tested, (i) the rate of decay of total IgG following infection declined with age and was driven consistently by the decrease in IgG3 and occasionally the decrease in IgG1; (ii) the proportion of IgG3 relative to IgG1 in the absence of infection increased with age; (iii) the increase in avidity index (the strength of association between the antibody and antigen) following infection was largely due to a rapid loss of non-avid compared to avid total IgG; and (iv) both avid and non-avid total IgG in the absence of infection increased with age. Further studies are required to understand the functional differences between IgG1 and IgG3 in order to determine their contribution to the longevity of protective immunity to malaria. Measuring changes in antibody avidity may be a better approach of detecting affinity maturation compared to avidity index due to the differential expansion and contraction of high and low avidity total IgG.
Collapse
Affiliation(s)
- Isaac Ssewanyana
- Infectious Diseases Research Collaboration, Kampala, Uganda.,Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John Rek
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Isabel Rodriguez
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Lindsey Wu
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Emmanuel Arinaitwe
- Infectious Diseases Research Collaboration, Kampala, Uganda.,Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Joaniter I Nankabirwa
- Infectious Diseases Research Collaboration, Kampala, Uganda.,School of Medicine, Makerere University, Kampala, Uganda
| | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | | | - Philip J Rosenthal
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Grant Dorsey
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda.,School of Medicine, Makerere University, Kampala, Uganda
| | - Chris Drakeley
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Bryan Greenhouse
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States.,Chan Zuckerberg Biohub, San Francisco, CA, United States
| | - Kevin K A Tetteh
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
44
|
Jelínková L, Jhun H, Eaton A, Petrovsky N, Zavala F, Chackerian B. An epitope-based malaria vaccine targeting the junctional region of circumsporozoite protein. NPJ Vaccines 2021; 6:13. [PMID: 33479242 PMCID: PMC7820318 DOI: 10.1038/s41541-020-00274-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/11/2020] [Indexed: 01/29/2023] Open
Abstract
A malaria vaccine that elicits long-lasting protection and is suitable for use in endemic areas remains urgently needed. Here, we assessed the immunogenicity and prophylactic efficacy of a vaccine targeting a recently described epitope on the major surface antigen on Plasmodium falciparum sporozoites, circumsporozoite protein (CSP). Using a virus-like particle (VLP)-based vaccine platform technology, we developed a vaccine that targets the junctional region between the N-terminal and central repeat regions of CSP. This region is recognized by monoclonal antibodies, including mAb CIS43, that have been shown to potently prevent liver invasion in animal models. We show that CIS43 VLPs elicit high-titer and long-lived anti-CSP antibody responses in mice and is immunogenic in non-human primates. In mice, vaccine immunogenicity was enhanced by using mixed adjuvant formulations. Immunization with CIS43 VLPs conferred partial protection from malaria infection in a mouse model, and passive transfer of serum from immunized macaques also inhibited parasite liver invasion in the mouse infection model. Our findings demonstrate that a Qβ VLP-based vaccine targeting the CIS43 epitope combined with various adjuvants is highly immunogenic in mice and macaques, elicits long-lasting anti-CSP antibodies, and inhibits parasite infection in a mouse model. Thus, the CIS43 VLP vaccine is a promising pre-erythrocytic malaria vaccine candidate.
Collapse
Affiliation(s)
- Lucie Jelínková
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Hugo Jhun
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Allison Eaton
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale, Adelaide, SA, 5046, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, SA, 5042, Australia
| | - Fidel Zavala
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Bryce Chackerian
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA.
| |
Collapse
|
45
|
Bell GJ, Agnandji ST, Asante KP, Ghansah A, Kamthunzi P, Emch M, Bailey JA. Impacts of Ecology, Parasite Antigenic Variation, and Human Genetics on RTS,S/AS01e Malaria Vaccine Efficacy. CURR EPIDEMIOL REP 2021; 8:79-88. [PMID: 34367877 PMCID: PMC8324449 DOI: 10.1007/s40471-021-00271-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Global malaria elimination has little chance of success without an effective vaccine. The first malaria vaccine, RTS,S/AS01e, demonstrated moderate efficacy against clinical malaria in phase III trials and is undergoing large-scale effectiveness trials in Africa. Importantly, the vaccine did not perform equally well between phase III study sites. Though reasons for the moderate efficacy and this variation are unclear, various mechanisms have been suggested. This review summarizes the recent literature on such mechanisms, with a focus on those involving landscape ecology, parasite antigenic variation, and human host genetic differences. RECENT FINDINGS Transmission intensity may have a role pre- and post-vaccination in modulating immune responses to the vaccine. Furthermore, malaria incidence may "rebound" in vaccinated populations living in high transmission intensity settings. There is growing evidence that both genetic variation in the parasite circumsporozoite protein and variation of human host genetic factors affect RTS,S vaccine efficacy. These genetic factors may be interacting in complex ways to produce variation in the natural and vaccine-induced immune responses that protect against malaria. SUMMARY Due to the modest efficacy of RTS,S/AS01e, the combinations of factors (ecological, parasite, human host) impacting its effectiveness must be clearly understood, as this information will be critical for implementation policy and future vaccine designs.
Collapse
Affiliation(s)
- Griffin J. Bell
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon ,Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | | | - Anita Ghansah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | | | - Michael Emch
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599 USA ,Department of Geography, University of North Carolina, Chapel Hill 220 E Cameron Ave, Chapel Hill, NC 27599 USA
| | - Jeffrey A. Bailey
- Department of Pathology and Laboratory Medicine, Brown University, 55 Claverick St, Rm 314B, Providence, RI 02912 USA
| |
Collapse
|
46
|
Suau R, Vidal M, Aguilar R, Ruiz-Olalla G, Vázquez-Santiago M, Jairoce C, Nhabomba AJ, Gyan B, Dosoo D, Asante KP, Owusu-Agyei S, Campo JJ, Izquierdo L, Cavanagh D, Coppel RL, Chauhan V, Angov E, Dutta S, Gaur D, Beeson JG, Moncunill G, Dobaño C. RTS,S/AS01 E malaria vaccine induces IgA responses against CSP and vaccine-unrelated antigens in African children in the phase 3 trial. Vaccine 2020; 39:687-698. [PMID: 33358704 DOI: 10.1016/j.vaccine.2020.12.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/05/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The evaluation of immune responses to RTS,S/AS01 has traditionally focused on immunoglobulin (Ig) G antibodies that are only moderately associated with protection. The role of other antibody isotypes that could also contribute to vaccine efficacy remains unclear. Here we investigated whether RTS,S/AS01E elicits antigen-specific serum IgA antibodies to the vaccine and other malaria antigens, and we explored their association with protection. METHODS Ninety-five children (age 5-17 months old at first vaccination) from the RTS,S/AS01E phase 3 clinical trial who received 3 doses of RTS,S/AS01E or a comparator vaccine were selected for IgA quantification 1 month post primary immunization. Two sites with different malaria transmission intensities (MTI) and clinical malaria cases and controls, were included. Measurements of IgA against different constructs of the circumsporozoite protein (CSP) vaccine antigen and 16 vaccine-unrelated Plasmodium falciparum antigens were performed using a quantitative suspension array assay. RESULTS RTS,S vaccination induced a 1.2 to 2-fold increase in levels of serum/plasma IgA antibodies to all CSP constructs, which was not observed upon immunization with a comparator vaccine. The IgA response against 13 out of 16 vaccine-unrelated P. falciparum antigens also increased after vaccination, and levels were higher in recipients of RTS,S than in comparators. IgA levels to malaria antigens before vaccination were more elevated in the high MTI than the low MTI site. No statistically significant association of IgA with protection was found in exploratory analyses. CONCLUSIONS RTS,S/AS01E induces IgA responses in peripheral blood against CSP vaccine antigens and other P. falciparum vaccine-unrelated antigens, similar to what we previously showed for IgG responses. Collectively, data warrant further investigation of the potential contribution of vaccine-induced IgA responses to efficacy and any possible interplay, either synergistic or antagonistic, with protective IgG, as identifying mediators of protection by RTS,S/AS01E immunization is necessary for the design of improved second-generation vaccines. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov: NCT008666191.
Collapse
Affiliation(s)
- Roger Suau
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Marta Vidal
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Gemma Ruiz-Olalla
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Miquel Vázquez-Santiago
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Chenjerai Jairoce
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929 Maputo, Mozambique.
| | - Augusto J Nhabomba
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929 Maputo, Mozambique
| | - Ben Gyan
- Noguchi Memorial Institute for Medical Research, University of Ghana, Ghana.
| | - David Dosoo
- Kintampo Health Research Centre, Kintampo, Ghana.
| | | | - Seth Owusu-Agyei
- Kintampo Health Research Centre, Kintampo, Ghana; Disease Control Department. London School of Hygiene and Tropical Medicine, London, UK
| | - Joseph J Campo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Luis Izquierdo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - 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, Edinburgh, UK.
| | - Ross L Coppel
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia.
| | - Virander Chauhan
- 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 (WRAIR), Silver Spring, MD, USA.
| | - Sheetij Dutta
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA.
| | - Deepak Gaur
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India; Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - James G Beeson
- Burnet Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Australia; Department of Medicine, University of Melbourne, Australia.
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929 Maputo, Mozambique.
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929 Maputo, Mozambique.
| |
Collapse
|
47
|
Kurtovic L, Boyle MJ, Beeson JG. Epitope masking may limit antibody boosting to malaria vaccines. Immunol Cell Biol 2020; 99:126-129. [PMID: 33152796 DOI: 10.1111/imcb.12415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
We discuss the study by McNamara et al., who report that low levels of antigen-specific antibodies in serum can limit the boosting of antibody and B-cell responses following immunization with live attenuated malaria sporozoites.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Burnet Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Michelle J Boyle
- Burnet Institute, Melbourne, VIC, Australia.,QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,Central Clinical School and Department of Microbiology, Monash University, Melbourne, VIC, Australia.,Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
48
|
Mo AXY, Pesce J, Augustine AD, Bodmer JL, Breen J, Leitner W, Hall BF. Understanding vaccine-elicited protective immunity against pre-erythrocytic stage malaria in endemic regions. Vaccine 2020; 38:7569-7577. [PMID: 33071001 DOI: 10.1016/j.vaccine.2020.09.071] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/26/2020] [Accepted: 09/24/2020] [Indexed: 12/17/2022]
Abstract
Recent malaria vaccine trials in endemic areas have yielded disparate results compared to studies conducted in non-endemic areas. A workshop was organized to discuss the differential pre-erythrocytic stage malaria vaccine (Pre-E-Vac) efficacies and underlying protective immunity under various conditions. It was concluded that many factors, including vaccine technology platforms, host genetics or physiologic conditions, and parasite and mosquito vector variations, may all contribute to Pre-E-Vac efficacy. Cross-disciplinary approaches are needed to decipher the multi-dimensional variables that contribute to the observed vaccine hypo-responsiveness. The malaria vaccine community has an opportunity to leverage recent advances in immunology, systems vaccinology, and high dimensionality data science methodologies to generate new clinical datasets with unprecedented levels of functional resolution as well as capitalize on existing datasets for comprehensive and aggregate analyses. These approaches would help to unlock our understanding of Pre-E-Vac immunology and to translate new candidates from the laboratory to the field more predictably.
Collapse
Affiliation(s)
- Annie X Y Mo
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA.
| | - John Pesce
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA
| | - Alison Deckhut Augustine
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA
| | | | - Joseph Breen
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA
| | - Wolfgang Leitner
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA
| | - B Fenton Hall
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA
| |
Collapse
|
49
|
Thompson HA, Hogan AB, Walker PGT, White MT, Cunnington AJ, Ockenhouse CF, Ghani AC. Modelling the roles of antibody titre and avidity in protection from Plasmodium falciparum malaria infection following RTS,S/AS01 vaccination. Vaccine 2020; 38:7498-7507. [PMID: 33041104 PMCID: PMC7607256 DOI: 10.1016/j.vaccine.2020.09.069] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 08/21/2020] [Accepted: 09/24/2020] [Indexed: 12/16/2022]
Abstract
Models capturing key malaria life-cycle stages can help us evaluate vaccine candidates. Model fitting revealed antibody avidity to be an important determinant of RTS,S vaccine efficacy. High avidity and titre were associated with increased levels of vaccine efficacy. Did not identify any thresholds of protection for either immune marker.
Anti-circumsporozoite antibody titres have been established as an essential indicator for evaluating the immunogenicity and protective capacity of the RTS,S/AS01 malaria vaccine. However, a new delayed-fractional dose regime of the vaccine was recently shown to increase vaccine efficacy, from 62.5% (95% CI 29.4–80.1%) under the original dosing schedule to 86.7% (95% CI, 66.8–94.6%) without a corresponding increase in antibody titres. Here we reanalyse the antibody data from this challenge trial to determine whether IgG avidity may help to explain efficacy better than IgG titre alone by adapting a within-host mathematical model of sporozoite inoculation. We demonstrate that a model incorporating titre and avidity provides a substantially better fit to the data than titre alone. These results also suggest that in individuals with a high antibody titre response that also show high avidity (both metrics in the top tercile of observed values) delayed-fractional vaccination provided near perfect protection upon first challenge (98.2% [95% Credible Interval 91.6–99.7%]). This finding suggests that the quality of the vaccine induced antibody response is likely to be an important determinant in the development of highly efficacious pre-erythrocytic vaccines against malaria.
Collapse
Affiliation(s)
- Hayley A Thompson
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom.
| | - Alexandra B Hogan
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Patrick G T Walker
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Michael T White
- Malaria: Parasites and Hosts, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| | | | | | - Azra C Ghani
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| |
Collapse
|
50
|
Abuga KM, Jones-Warner W, Hafalla JCR. Immune responses to malaria pre-erythrocytic stages: Implications for vaccine development. Parasite Immunol 2020; 43:e12795. [PMID: 32981095 PMCID: PMC7612353 DOI: 10.1111/pim.12795] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/26/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022]
Abstract
Radiation-attenuated sporozoites induce sterilizing immunity and remain the 'gold standard' for malaria vaccine development. Despite practical challenges in translating these whole sporozoite vaccines to large-scale intervention programmes, they have provided an excellent platform to dissect the immune responses to malaria pre-erythrocytic (PE) stages, comprising both sporozoites and exoerythrocytic forms. Investigations in rodent models have provided insights that led to the clinical translation of various vaccine candidates-including RTS,S/AS01, the most advanced candidate currently in a trial implementation programme in three African countries. With advances in immunology, transcriptomics and proteomics, and application of lessons from past failures, an effective, long-lasting and wide-scale malaria PE vaccine remains feasible. This review underscores the progress in PE vaccine development, focusing on our understanding of host-parasite immunological crosstalk in the tissue environments of the skin and the liver. We highlight possible gaps in the current knowledge of PE immunity that can impact future malaria vaccine development efforts.
Collapse
Affiliation(s)
- Kelvin Mokaya Abuga
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK.,Department of Epidemiology and Demography, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - William Jones-Warner
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Julius Clemence R Hafalla
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|