1
|
Duffy PE, Gorres JP, Healy SA, Fried M. Malaria vaccines: a new era of prevention and control. Nat Rev Microbiol 2024:10.1038/s41579-024-01065-7. [PMID: 39025972 DOI: 10.1038/s41579-024-01065-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 07/20/2024]
Abstract
Malaria killed over 600,000 people in 2022, a death toll that has not improved since 2015. Additionally, parasites and mosquitoes resistant to existing interventions are spreading across Africa and other regions. Vaccines offer hope to reduce the mortality burden: the first licensed malaria vaccines, RTS,S and R21, will be widely deployed in 2024 and should substantially reduce childhood deaths. In this Review, we provide an overview of the malaria problem and the Plasmodium parasite, then describe the RTS,S and R21 vaccines (the first vaccines for any human parasitic disease), summarizing their benefits and limitations. We explore next-generation vaccines designed using new knowledge of malaria pathogenesis and protective immunity, which incorporate antigens and platforms to elicit effective immune responses against different parasite stages in human or mosquito hosts. We describe a decision-making process that prioritizes malaria vaccine candidates for development in a resource-constrained environment. Future vaccines might improve upon the protective efficacy of RTS,S or R21 for children, or address the wider malaria scourge by preventing pregnancy malaria, reducing the burden of Plasmodium vivax or accelerating malaria elimination.
Collapse
Affiliation(s)
- Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - J Patrick Gorres
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Obaldía N, Da Silva Filho JL, Núñez M, Glass KA, Oulton T, Achcar F, Wirjanata G, Duraisingh M, Felgner P, Tetteh KK, Bozdech Z, Otto TD, Marti M. Sterile protection against P. vivax malaria by repeated blood stage infection in the Aotus monkey model. Life Sci Alliance 2024; 7:e202302524. [PMID: 38158220 PMCID: PMC10756917 DOI: 10.26508/lsa.202302524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024] Open
Abstract
The malaria parasite Plasmodium vivax remains a major global public health challenge, and no vaccine is approved for use in humans. Here, we assessed whether P. vivax strain-transcendent immunity can be achieved by repeated infection in Aotus monkeys. Sterile immunity was achieved after two homologous infections, whereas subsequent heterologous challenge provided only partial protection. IgG levels based on P. vivax lysate ELISA and protein microarray increased with repeated infections and correlated with the level of homologous protection. Parasite transcriptional profiles provided no evidence of major antigenic switching upon homologous or heterologous challenge. However, we observed significant sequence diversity and transcriptional differences in the P. vivax core gene repertoire between the two strains used in the study, suggesting that partial protection upon heterologous challenge is due to molecular differences between strains rather than immune evasion by antigenic switching. Our study demonstrates that sterile immunity against P. vivax can be achieved by repeated homologous blood stage infection in Aotus monkeys, thus providing a benchmark to test the efficacy of candidate blood stage P. vivax malaria vaccines.
Collapse
Affiliation(s)
- Nicanor Obaldía
- Departamento de Investigaciones en Parasitologia, Instituto Conmemorativo Gorgas de Estudios de la Salud, Panamá City, Republic of Panamá
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Harvard University, Boston, MA, USA
- https://ror.org/00vtgdb53 Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Joao Luiz Da Silva Filho
- https://ror.org/00vtgdb53 Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- https://ror.org/02crff812 Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Marlon Núñez
- Departamento de Investigaciones en Parasitologia, Instituto Conmemorativo Gorgas de Estudios de la Salud, Panamá City, Republic of Panamá
| | - Katherine A Glass
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Tate Oulton
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Fiona Achcar
- https://ror.org/00vtgdb53 Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- https://ror.org/02crff812 Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
| | - Grennady Wirjanata
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Manoj Duraisingh
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Philip Felgner
- Institute for Immunology, University of California, Irvine, CA, USA
| | - Kevin Ka Tetteh
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Thomas D Otto
- https://ror.org/00vtgdb53 Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Matthias Marti
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Harvard University, Boston, MA, USA
- https://ror.org/00vtgdb53 Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- https://ror.org/02crff812 Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Thomson-Luque R, Stabler TC, Fürle K, Silva JC, Daubenberger C. Plasmodium falciparum merozoite surface protein 1 as asexual blood stage malaria vaccine candidate. Expert Rev Vaccines 2024; 23:160-173. [PMID: 38100310 DOI: 10.1080/14760584.2023.2295430] [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: 09/25/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION Malaria represents a public health challenge in tropical and subtropical regions, and currently deployed control strategies are likely insufficient to drive elimination of malaria. Development and improvement of malaria vaccines might be key to reduce disease burden. Vaccines targeting asexual blood stages of the parasite have shown limited efficacy when studied in human trials conducted over the past decades. AREAS COVERED Vaccine candidates based on the merozoite surface protein 1 (MSP1) were initially envisioned as one of the most promising approaches to provide immune protection against asexual blood-stage malaria. Successful immunization studies in monkey involved the use of the full-length MSP1 (MSP1FL) as vaccine construct. Vaccines using MSP1FL for immunization have the potential benefit of including numerous conserved B-cell and T-cell epitopes. This could result in improved parasite strain-transcending, protective immunity in the field. We review outcomes of clinical trials that utilized a variety of MSP1 constructs and formulations, including MSP1FL, either alone or in combination with other antigens, in both animal models and humans. EXPERT OPINION Novel approaches to analyze breadth and magnitude of effector functions of MSP1-targeting antibodies in volunteers undergoing experimental vaccination and controlled human malaria infection will help to define correlates of protective immunity.
Collapse
Affiliation(s)
- Richard Thomson-Luque
- Centre for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany
- Sumaya-Biotech GmbH & Co. KG Heidelberg, Germany
| | - Thomas C Stabler
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- University of Basel Basel, Switzerland
- Swiss Tropical and Public Health Institute Allschwil, Switzerland
| | - Kristin Fürle
- Centre for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Joana C Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa (GHTM IHMT, UNL), Lisbon, Portugal
| | - Claudia Daubenberger
- University of Basel Basel, Switzerland
- Swiss Tropical and Public Health Institute Allschwil, Switzerland
| |
Collapse
|
4
|
Silk SE, Kalinga WF, Mtaka IM, Lilolime NS, Mpina M, Milando F, Ahmed S, Diouf A, Mkwepu F, Simon B, Athumani T, Rashid M, Mohammed L, Lweno O, Ali AM, Nyaulingo G, Mwalimu B, Mswata S, Mwamlima TG, Barrett JR, Wang LT, Themistocleous Y, King LDW, Hodgson SH, Payne RO, Nielsen CM, Lawrie AM, Nugent FL, Cho JS, Long CA, Miura K, Draper SJ, Minassian AM, Olotu AI. Superior antibody immunogenicity of a viral-vectored RH5 blood-stage malaria vaccine in Tanzanian infants as compared to adults. MED 2023; 4:668-686.e7. [PMID: 37572659 DOI: 10.1016/j.medj.2023.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/23/2023] [Accepted: 07/11/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND RH5 is a leading blood-stage candidate antigen for a Plasmodium falciparum vaccine; however, its safety and immunogenicity in malaria-endemic populations are unknown. METHODS A phase 1b, single-center, dose-escalation, age-de-escalation, double-blind, randomized, controlled trial was conducted in Bagamoyo, Tanzania (NCT03435874). Between 12th April and 25th October 2018, 63 healthy adults (18-35 years), young children (1-6 years), and infants (6-11 months) received a priming dose of viral-vectored ChAd63 RH5 or rabies control vaccine. Sixty participants were boosted with modified vaccinia virus Ankara (MVA) RH5 or rabies control vaccine 8 weeks later and completed 6 months of follow-up post priming. Primary outcomes were the number of solicited and unsolicited adverse events post vaccination and the number of serious adverse events over the study period. Secondary outcomes included measures of the anti-RH5 immune response. FINDINGS Vaccinations were well tolerated, with profiles comparable across groups. No serious adverse events were reported. Vaccination induced RH5-specific cellular and humoral responses. Higher anti-RH5 serum immunoglobulin G (IgG) responses were observed post boost in young children and infants compared to adults. Vaccine-induced antibodies showed growth inhibition activity (GIA) in vitro against P. falciparum blood-stage parasites; their highest levels were observed in infants. CONCLUSIONS The ChAd63-MVA RH5 vaccine shows acceptable safety and reactogenicity and encouraging immunogenicity in children and infants residing in a malaria-endemic area. The levels of functional GIA observed in RH5-vaccinated infants are the highest reported to date following human vaccination. These data support onward clinical development of RH5-based blood-stage vaccines to protect against clinical malaria in young African infants. FUNDING Medical Research Council, London, UK.
Collapse
Affiliation(s)
- Sarah E Silk
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Wilmina F Kalinga
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Ivanny M Mtaka
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Nasoro S Lilolime
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Maximillian Mpina
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Florence Milando
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Saumu Ahmed
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Fatuma Mkwepu
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Beatus Simon
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Thabit Athumani
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Mohammed Rashid
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Latipha Mohammed
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Omary Lweno
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Ali M Ali
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Gloria Nyaulingo
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Bakari Mwalimu
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Sarah Mswata
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Tunu G Mwamlima
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Jordan R Barrett
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Lawrence T Wang
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Yrene Themistocleous
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Lloyd D W King
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Susanne H Hodgson
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Ruth O Payne
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Carolyn M Nielsen
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Alison M Lawrie
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Fay L Nugent
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Jee-Sun Cho
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Angela M Minassian
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Ally I Olotu
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| |
Collapse
|
5
|
Miura K, Diouf A, Fay MP, Barrett JR, Payne RO, Olotu AI, Minassian AM, Silk SE, Draper SJ, Long CA. Assessment of precision in growth inhibition assay (GIA) using human anti-PfRH5 antibodies. Malar J 2023; 22:159. [PMID: 37208733 PMCID: PMC10196285 DOI: 10.1186/s12936-023-04591-6] [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: 02/14/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND For blood-stage malaria vaccine development, the in vitro growth inhibition assay (GIA) has been widely used to evaluate functionality of vaccine-induced antibodies (Ab), and Plasmodium falciparum reticulocyte-binding protein homolog 5 (RH5) is a leading blood-stage antigen. However, precision, also called "error of assay (EoA)", in GIA readouts and the source of EoA has not been evaluated systematically. METHODS In the Main GIA experiment, 4 different cultures of P. falciparum 3D7 parasites were prepared with red blood cells (RBC) collected from 4 different donors. For each culture, 7 different anti-RH5 Ab (either monoclonal or polyclonal Ab) were tested by GIA at two concentrations on three different days (168 data points). To evaluate sources of EoA in % inhibition in GIA (%GIA), a linear model fit was conducted including donor (source of RBC) and day of GIA as independent variables. In addition, 180 human anti-RH5 polyclonal Ab were tested in a Clinical GIA experiment, where each Ab was tested at multiple concentrations in at least 3 independent GIAs using different RBCs (5,093 data points). The standard deviation (sd) in %GIA and in GIA50 (Ab concentration that gave 50%GIA) readouts, and impact of repeat assays on 95% confidence interval (95%CI) of these readouts was estimated. RESULTS The Main GIA experiment revealed that the RBC donor effect was much larger than the day effect, and an obvious donor effect was also observed in the Clinical GIA experiment. Both %GIA and log-transformed GIA50 data reasonably fit a constant sd model, and sd of %GIA and log-transformed GIA50 measurements were calculated as 7.54 and 0.206, respectively. Taking the average of three repeat assays (using three different RBCs) reduces the 95%CI width in %GIA or in GIA50 measurements by ~ half compared to a single assay. CONCLUSIONS The RBC donor effect (donor-to-donor variance on the same day) in GIA was much bigger than the day effect (day-to-day variance using the same donor's RBC) at least for the RH5 Ab evaluated in this study; thus, future GIA studies should consider the donor effect. In addition, the 95%CI for %GIA and GIA50 shown here help when comparing GIA results from different samples/groups/studies; therefore, this study supports future malaria blood-stage vaccine development.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA.
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Michael P Fay
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Jordan R Barrett
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Ruth O Payne
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Ally I Olotu
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Angela M Minassian
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Sarah E Silk
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| |
Collapse
|
6
|
Bjerkan L, Visweswaran GRR, Gudjonsson A, Labbé GM, Quinkert D, Pattinson DJ, Spång HCL, Draper SJ, Bogen B, Braathen R. APC-Targeted DNA Vaccination Against Reticulocyte-Binding Protein Homolog 5 Induces Plasmodium falciparum-Specific Neutralizing Antibodies and T Cell Responses. Front Immunol 2021; 12:720550. [PMID: 34733274 PMCID: PMC8558525 DOI: 10.3389/fimmu.2021.720550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/30/2021] [Indexed: 11/20/2022] Open
Abstract
Targeted delivery of antigen to antigen presenting cells (APCs) is an efficient way to induce robust antigen-specific immune responses. Here, we present a novel DNA vaccine that targets the Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5), a leading blood-stage antigen of the human malaria pathogen, to APCs. The vaccine is designed as bivalent homodimers where each chain is composed of an amino-terminal single chain fragment variable (scFv) targeting unit specific for major histocompatibility complex class II (MHCII) expressed on APCs, and a carboxyl-terminal antigenic unit genetically linked by the dimerization unit. This vaccine format, named “Vaccibody”, has previously been successfully applied for antigens from other infectious diseases including influenza and HIV, as well as for tumor antigens. Recently, the crystal structure and key functional antibody epitopes for the truncated version of PfRH5 (PfRH5ΔNL) were characterized, suggesting PfRH5ΔNL to be a promising candidate for next-generation PfRH5 vaccine design. In this study, we explored the APC-targeting strategy for a PfRH5ΔNL-containing DNA vaccine. BALB/c mice immunized with the targeted vaccine induced higher PfRH5-specific IgG1 antibody responses than those vaccinated with a non-targeted vaccine or antigen alone. The APC-targeted vaccine also efficiently induced rapid IFN-γ and IL-4 T cell responses. Furthermore, the vaccine-induced PfRH5-specific IgG showed inhibition of growth of the P. falciparum 3D7 clone parasite in vitro. Finally, sera obtained after vaccination with this targeted vaccine competed for the same epitopes as PfRH5-specific mAbs from vaccinated humans. Robust humoral responses were also induced by a similar P. vivax Duffy-binding protein (PvDBP)-containing targeted DNA vaccine. Our data highlight a novel targeted vaccine platform for the development of vaccines against blood-stage malaria.
Collapse
Affiliation(s)
- Louise Bjerkan
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Arnar Gudjonsson
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Doris Quinkert
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Heidi C L Spång
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Simon J Draper
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Bjarne Bogen
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ranveig Braathen
- Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
7
|
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: 14] [Impact Index Per Article: 4.7] [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
|
8
|
Minassian AM, Silk SE, Barrett JR, Nielsen CM, Miura K, Diouf A, Loos C, Fallon JK, Michell AR, White MT, Edwards NJ, Poulton ID, Mitton CH, Payne RO, Marks M, Maxwell-Scott H, Querol-Rubiera A, Bisnauthsing K, Batra R, Ogrina T, Brendish NJ, Themistocleous Y, Rawlinson TA, Ellis KJ, Quinkert D, Baker M, Lopez Ramon R, Ramos Lopez F, Barfod L, Folegatti PM, Silman D, Datoo M, Taylor IJ, Jin J, Pulido D, Douglas AD, de Jongh WA, Smith R, Berrie E, Noe AR, Diggs CL, Soisson LA, Ashfield R, Faust SN, Goodman AL, Lawrie AM, Nugent FL, Alter G, Long CA, Draper SJ. Reduced blood-stage malaria growth and immune correlates in humans following RH5 vaccination. MED 2021; 2:701-719.e19. [PMID: 34223402 PMCID: PMC8240500 DOI: 10.1016/j.medj.2021.03.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/19/2021] [Accepted: 03/25/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Development of an effective vaccine against the pathogenic blood-stage infection of human malaria has proved challenging, and no candidate vaccine has affected blood-stage parasitemia following controlled human malaria infection (CHMI) with blood-stage Plasmodium falciparum. METHODS We undertook a phase I/IIa clinical trial in healthy adults in the United Kingdom of the RH5.1 recombinant protein vaccine, targeting the P. falciparum reticulocyte-binding protein homolog 5 (RH5), formulated in AS01B adjuvant. We assessed safety, immunogenicity, and efficacy against blood-stage CHMI. Trial registered at ClinicalTrials.gov, NCT02927145. FINDINGS The RH5.1/AS01B formulation was administered using a range of RH5.1 protein vaccine doses (2, 10, and 50 μg) and was found to be safe and well tolerated. A regimen using a delayed and fractional third dose, in contrast to three doses given at monthly intervals, led to significantly improved antibody response longevity over ∼2 years of follow-up. Following primary and secondary CHMI of vaccinees with blood-stage P. falciparum, a significant reduction in parasite growth rate was observed, defining a milestone for the blood-stage malaria vaccine field. We show that growth inhibition activity measured in vitro using purified immunoglobulin G (IgG) antibody strongly correlates with in vivo reduction of the parasite growth rate and also identify other antibody feature sets by systems serology, including the plasma anti-RH5 IgA1 response, that are associated with challenge outcome. CONCLUSIONS Our data provide a new framework to guide rational design and delivery of next-generation vaccines to protect against malaria disease. FUNDING This study was supported by USAID, UK MRC, Wellcome Trust, NIAID, and the NIHR Oxford-BRC.
Collapse
Affiliation(s)
| | - Sarah E. Silk
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | | | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Carolin Loos
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Ashlin R. Michell
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Michael T. White
- Department of Parasites and Insect Vectors, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Nick J. Edwards
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Ian D. Poulton
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Celia H. Mitton
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Ruth O. Payne
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Michael Marks
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Hector Maxwell-Scott
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Antonio Querol-Rubiera
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Karen Bisnauthsing
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Rahul Batra
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | - Tatiana Ogrina
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Nathan J. Brendish
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | | | | | | | - Doris Quinkert
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Megan Baker
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | | | - Lea Barfod
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Daniel Silman
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Mehreen Datoo
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Iona J. Taylor
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Jing Jin
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - David Pulido
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Willem A. de Jongh
- ExpreSion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, Hørsholm 2970, Denmark
| | - Robert Smith
- Clinical BioManufacturing Facility, University of Oxford, Oxford OX3 7JT, UK
| | - Eleanor Berrie
- Clinical BioManufacturing Facility, University of Oxford, Oxford OX3 7JT, UK
| | | | | | | | | | - Saul N. Faust
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Anna L. Goodman
- Centre for Clinical Infection and Diagnostics Research, King’s College London and Guy’s & St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK
| | | | - Fay L. Nugent
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Galit Alter
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
9
|
Tanner R, Hoogkamer E, Bitencourt J, White A, Boot C, Sombroek CC, Harris SA, O'Shea MK, Wright D, Wittenberg R, Sarfas C, Satti I, Verreck FA, Sharpe SA, Fletcher HA, McShane H. The in vitro direct mycobacterial growth inhibition assay (MGIA) for the early evaluation of TB vaccine candidates and assessment of protective immunity: a protocol for non-human primate cells. F1000Res 2021; 10:257. [PMID: 33976866 PMCID: PMC8097740 DOI: 10.12688/f1000research.51640.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 04/04/2024] Open
Abstract
The only currently available approach to early efficacy testing of tuberculosis (TB) vaccine candidates is in vivo preclinical challenge models. These typically include mice, guinea pigs and non-human primates (NHPs), which must be exposed to virulent M.tb in a 'challenge' experiment following vaccination in order to evaluate protective efficacy. This procedure results in disease development and is classified as 'Moderate' in severity under EU legislation and UK ASPA licensure. Furthermore, experiments are relatively long and animals must be maintained in high containment level facilities, making them relatively costly. We describe an in vitro protocol for the direct mycobacterial growth inhibition assay (MGIA) for use in the macaque model of TB vaccine development with the aim of overcoming some of these limitations. Importantly, using an in vitro assay in place of in vivo M.tb challenge represents a significant refinement to the existing procedure for early vaccine efficacy testing. Peripheral blood mononuclear cell and autologous serum samples collected from vaccinated and unvaccinated control animals are co-cultured with mycobacteria in a 48-well plate format for 96 hours. Adherent monocytes are then lysed to release intracellular mycobacteria which is quantified using the BACTEC MGIT system and colony-forming units determined relative to an inoculum control and stock standard curve. We discuss related optimisation and characterisation experiments, and review evidence that the direct NHP MGIA provides a biologically relevant model of vaccine-induced protection. The potential end-users of the NHP MGIA are academic and industry organisations that conduct the assessment of TB vaccine candidates and associated protective immunity using the NHP model. This approach aims to provide a method for high-throughput down-selection of vaccine candidates going forward to in vivo efficacy testing, thus expediting the development of a more efficacious TB vaccine and offering potential refinement and reduction to the use of NHPs for this purpose.
Collapse
Affiliation(s)
- Rachel Tanner
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| | - Emily Hoogkamer
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
- Public Health England, Salisbury, SP4 0JG, UK
| | - Julia Bitencourt
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
- Gonҫalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, 40296-710, Brazil
| | | | - Charelle Boot
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, 2288 GJ, The Netherlands
| | - Claudia C. Sombroek
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, 2288 GJ, The Netherlands
| | | | - Matthew K. O'Shea
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, UK, Birmingham, B15 2TH, UK
| | - Daniel Wright
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| | - Rachel Wittenberg
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| | | | - Iman Satti
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| | - Frank A.W. Verreck
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, 2288 GJ, The Netherlands
| | | | - Helen A. Fletcher
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
- London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Helen McShane
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| |
Collapse
|
10
|
Tanner R, Hoogkamer E, Bitencourt J, White A, Boot C, Sombroek CC, Harris SA, O'Shea MK, Wright D, Wittenberg R, Sarfas C, Satti I, Verreck FAW, Sharpe SA, Fletcher HA, McShane H. The in vitro direct mycobacterial growth inhibition assay (MGIA) for the early evaluation of TB vaccine candidates and assessment of protective immunity: a protocol for non-human primate cells. F1000Res 2021; 10:257. [PMID: 33976866 PMCID: PMC8097740.2 DOI: 10.12688/f1000research.51640.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/15/2021] [Indexed: 11/29/2022] Open
Abstract
The only currently available approach to early efficacy testing of tuberculosis (TB) vaccine candidates is in vivo preclinical challenge models. These typically include mice, guinea pigs and non-human primates (NHPs), which must be exposed to virulent M.tb in a 'challenge' experiment following vaccination in order to evaluate protective efficacy. This procedure results in disease development and is classified as 'Moderate' in severity under EU legislation and UK ASPA licensure. Furthermore, experiments are relatively long and animals must be maintained in high containment level facilities, making them relatively costly. We describe an in vitro protocol for the direct mycobacterial growth inhibition assay (MGIA) for use in the macaque model of TB vaccine development with the aim of overcoming some of these limitations. Importantly, using an in vitro assay in place of in vivo M.tb challenge represents a significant refinement to the existing procedure for early vaccine efficacy testing. Peripheral blood mononuclear cell and autologous serum samples collected from vaccinated and unvaccinated control animals are co-cultured with mycobacteria in a 48-well plate format for 96 hours. Adherent monocytes are then lysed to release intracellular mycobacteria which is quantified using the BACTEC MGIT system and colony-forming units determined relative to an inoculum control and stock standard curve. We discuss related optimisation and characterisation experiments, and review evidence that the direct NHP MGIA provides a biologically relevant model of vaccine-induced protection. The potential end-users of the NHP MGIA are academic and industry organisations that conduct the assessment of TB vaccine candidates and associated protective immunity using the NHP model. This approach aims to provide a method for high-throughput down-selection of vaccine candidates going forward to in vivo efficacy testing, thus expediting the development of a more efficacious TB vaccine and offering potential refinement and reduction to the use of NHPs for this purpose.
Collapse
Affiliation(s)
- Rachel Tanner
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| | - Emily Hoogkamer
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
- Public Health England, Salisbury, SP4 0JG, UK
| | - Julia Bitencourt
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
- Gonҫalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, 40296-710, Brazil
| | | | - Charelle Boot
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, 2288 GJ, The Netherlands
| | - Claudia C Sombroek
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, 2288 GJ, The Netherlands
| | - Stephanie A Harris
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| | - Matthew K O'Shea
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, UK, Birmingham, B15 2TH, UK
| | - Daniel Wright
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| | - Rachel Wittenberg
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| | | | - Iman Satti
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| | - Frank A W Verreck
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, 2288 GJ, The Netherlands
| | | | - Helen A Fletcher
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
- London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Helen McShane
- Nuffield Department of Medicine, The Jenner Institute, Oxford, OX3 7DQ, UK
| |
Collapse
|
11
|
Flynn O, Dillane K, Lanza JS, Marshall JM, Jin J, Silk SE, Draper SJ, Moore AC. Low Adenovirus Vaccine Doses Administered to Skin Using Microneedle Patches Induce Better Functional Antibody Immunogenicity as Compared to Systemic Injection. Vaccines (Basel) 2021; 9:vaccines9030299. [PMID: 33810085 PMCID: PMC8005075 DOI: 10.3390/vaccines9030299] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/23/2021] [Accepted: 02/27/2021] [Indexed: 01/02/2023] Open
Abstract
Adenovirus-based vaccines are demonstrating promising clinical potential for multiple infectious diseases, including COVID-19. However, the immunogenicity of the vector itself decreases its effectiveness as a boosting vaccine due to the induction of strong anti-vector neutralizing immunity. Here we determined how dissolvable microneedle patches (DMN) for skin immunization can overcome this issue, using a clinically-relevant adenovirus-based Plasmodium falciparum malaria vaccine, AdHu5–PfRH5, in mice. Incorporation of vaccine into patches significantly enhanced its thermostability compared to the liquid form. Conventional high dose repeated immunization by the intramuscular (IM) route induced low antigen-specific IgG titres and high anti-vector immunity. A low priming dose of vaccine, by the IM route, but more so using DMN patches, induced the most efficacious immune responses, assessed by parasite growth inhibitory activity (GIA) assays. Administration of low dose AdHu5–PfRH5 using patches to the skin, boosted by high dose IM, induced the highest antigen-specific serum IgG response after boosting, the greatest skewing of the antibody response towards the antigen and away from the vector, and the highest efficacy. This study therefore demonstrates that repeated use of the same adenovirus vaccine can be highly immunogenic towards the transgene if a low dose is used to prime the response. It also provides a method of stabilizing adenovirus vaccine, in easy-to-administer dissolvable microneedle patches, permitting storage and distribution out of cold chain.
Collapse
Affiliation(s)
- Olivia Flynn
- School of Pharmacy, University College Cork, T12 XF62 Cork, Ireland; (O.F.); (K.D.); (J.S.L.)
| | - Kate Dillane
- School of Pharmacy, University College Cork, T12 XF62 Cork, Ireland; (O.F.); (K.D.); (J.S.L.)
| | - Juliane Sousa Lanza
- School of Pharmacy, University College Cork, T12 XF62 Cork, Ireland; (O.F.); (K.D.); (J.S.L.)
| | - Jennifer M. Marshall
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (J.M.M.); (J.J.); (S.E.S.); (S.J.D.)
| | - Jing Jin
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (J.M.M.); (J.J.); (S.E.S.); (S.J.D.)
| | - Sarah E. Silk
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (J.M.M.); (J.J.); (S.E.S.); (S.J.D.)
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (J.M.M.); (J.J.); (S.E.S.); (S.J.D.)
| | - Anne C. Moore
- School of Pharmacy, University College Cork, T12 XF62 Cork, Ireland; (O.F.); (K.D.); (J.S.L.)
- School of Biochemistry and Cell Biology, University College Cork, T12 XF62 Cork, Ireland
- Correspondence:
| |
Collapse
|
12
|
Schneider CG, Taylor JA, Sibilo MQ, Miura K, Mallory KL, Mann C, Karch C, Beck Z, Matyas GR, Long CA, Bergmann-Leitner E, Burkhard P, Angov E. Orientation of Antigen Display on Self-Assembling Protein Nanoparticles Influences Immunogenicity. Vaccines (Basel) 2021; 9:vaccines9020103. [PMID: 33572803 PMCID: PMC7911071 DOI: 10.3390/vaccines9020103] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 11/16/2022] Open
Abstract
Self-assembling protein nanoparticles (SAPN) serve as a repetitive antigen delivery platform with high-density epitope display; however, antigen characteristics such as size and epitope presentation can influence the immunogenicity of the assembled particle and are aspects to consider for a rationally designed effective vaccine. Here, we characterize the folding and immunogenicity of heterogeneous antigen display by integrating (a) dual-stage antigen SAPN presenting the P. falciparum (Pf) merozoite surface protein 1 subunit, PfMSP119, and Pf cell-traversal protein for ookinetes and sporozoites, PfCelTOS, in addition to (b) a homogenous antigen SAPN displaying two copies of PfCelTOS. Mice and rabbits were utilized to evaluate antigen-specific humoral and cellular induction as well as functional antibodies via growth inhibition of the blood-stage parasite. We demonstrate that antigen orientation and folding influence the elicited immune response, and when appropriately designed, SAPN can serve as an adaptable platform for an effective multi-antigen display.
Collapse
Affiliation(s)
- Cosette G. Schneider
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (C.G.S.); (J.A.T.); (M.Q.S.); (K.L.M.); (C.M.); (E.B.-L.)
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37831, USA
| | - Justin A. Taylor
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (C.G.S.); (J.A.T.); (M.Q.S.); (K.L.M.); (C.M.); (E.B.-L.)
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37831, USA
| | - Michael Q. Sibilo
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (C.G.S.); (J.A.T.); (M.Q.S.); (K.L.M.); (C.M.); (E.B.-L.)
- Parsons Corporation, Centreville, VA 20120, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20892, USA; (K.M.); (C.A.L.)
| | - Katherine L. Mallory
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (C.G.S.); (J.A.T.); (M.Q.S.); (K.L.M.); (C.M.); (E.B.-L.)
- Parsons Corporation, Centreville, VA 20120, USA
| | - Christopher Mann
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (C.G.S.); (J.A.T.); (M.Q.S.); (K.L.M.); (C.M.); (E.B.-L.)
- Parsons Corporation, Centreville, VA 20120, USA
| | - Christopher Karch
- Laboratory of Antigen and Adjuvants, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (C.K.); (Z.B.); (G.R.M.)
- Henry Jackson Foundation, Bethesda, MD 20817, USA
| | - Zoltan Beck
- Laboratory of Antigen and Adjuvants, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (C.K.); (Z.B.); (G.R.M.)
- Henry Jackson Foundation, Bethesda, MD 20817, USA
| | - Gary R. Matyas
- Laboratory of Antigen and Adjuvants, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (C.K.); (Z.B.); (G.R.M.)
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20892, USA; (K.M.); (C.A.L.)
| | - Elke Bergmann-Leitner
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (C.G.S.); (J.A.T.); (M.Q.S.); (K.L.M.); (C.M.); (E.B.-L.)
| | | | - Evelina Angov
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (C.G.S.); (J.A.T.); (M.Q.S.); (K.L.M.); (C.M.); (E.B.-L.)
- Correspondence: ; Tel.: +1-301-319-9614
| |
Collapse
|
13
|
Good MF, Stanisic DI. Whole parasite vaccines for the asexual blood stages ofPlasmodium. Immunol Rev 2019; 293:270-282. [DOI: 10.1111/imr.12819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Michael F. Good
- Institute for Glycomics Griffith University Gold Coast Qld. Australia
| | | |
Collapse
|
14
|
Boyle MJ, Chan JA, Handayuni I, Reiling L, Feng G, Hilton A, Kurtovic L, Oyong D, Piera KA, Barber BE, William T, Eisen DP, Minigo G, Langer C, Drew DR, de Labastida Rivera F, Amante FH, Williams TN, Kinyanjui S, Marsh K, Doolan DL, Engwerda C, Fowkes FJI, Grigg MJ, Mueller I, McCarthy JS, Anstey NM, Beeson JG. IgM in human immunity to Plasmodium falciparum malaria. SCIENCE ADVANCES 2019; 5:eaax4489. [PMID: 31579826 PMCID: PMC6760923 DOI: 10.1126/sciadv.aax4489] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 09/03/2019] [Indexed: 05/15/2023]
Abstract
Most studies on human immunity to malaria have focused on the roles of immunoglobulin G (IgG), whereas the roles of IgM remain undefined. Analyzing multiple human cohorts to assess the dynamics of malaria-specific IgM during experimentally induced and naturally acquired malaria, we identified IgM activity against blood-stage parasites. We found that merozoite-specific IgM appears rapidly in Plasmodium falciparum infection and is prominent during malaria in children and adults with lifetime exposure, together with IgG. Unexpectedly, IgM persisted for extended periods of time; we found no difference in decay of merozoite-specific IgM over time compared to that of IgG. IgM blocked merozoite invasion of red blood cells in a complement-dependent manner. IgM was also associated with significantly reduced risk of clinical malaria in a longitudinal cohort of children. These findings suggest that merozoite-specific IgM is an important functional and long-lived antibody response targeting blood-stage malaria parasites that contributes to malaria immunity.
Collapse
Affiliation(s)
- M. J. Boyle
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Corresponding author. (M.J.B.); (J.G.B.)
| | - J. A. Chan
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - I. Handayuni
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - L. Reiling
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - G. Feng
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - A. Hilton
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - L. Kurtovic
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | - D. Oyong
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Charles Darwin University, Darwin, Northern Territory, Australia
| | - K. A. Piera
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - B. E. Barber
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
| | - T. William
- Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
- Gleneagles Hospital Kota Kinabalu Sabah, Malaysia
| | - D. P. Eisen
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - G. Minigo
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Charles Darwin University, Darwin, Northern Territory, Australia
| | - C. Langer
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - D. R. Drew
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | | | - F. H. Amante
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - T. N. Williams
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Imperial College, London, UK
| | - S. Kinyanjui
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - K. Marsh
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - D. L. Doolan
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - C. Engwerda
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - F. J. I. Fowkes
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Epidemiology and Preventive Medicine, Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
| | - M. J. Grigg
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
| | - I. Mueller
- Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Parasites and Insect Vectors, Institute Pasteur, Paris, France
| | - J. S. McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- The University of Queensland, Brisbane, Queensland, Australia
| | - N. M. Anstey
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Charles Darwin University, Darwin, Northern Territory, Australia
| | - J. G. Beeson
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Department of Microbiology and Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Corresponding author. (M.J.B.); (J.G.B.)
| |
Collapse
|
15
|
Elizardez YB, Fotoran WL, Junior AJG, Curado I, Junior NK, Monteiro EF, Romero Neto I, Wunderlich G, Kirchgatter K. Recombinant proteins of Plasmodium malariae merozoite surface protein 1 (PmMSP1): Testing immunogenicity in the BALB/c model and potential use as diagnostic tool. PLoS One 2019; 14:e0219629. [PMID: 31344067 PMCID: PMC6657842 DOI: 10.1371/journal.pone.0219629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/27/2019] [Indexed: 01/06/2023] Open
Abstract
Background Plasmodium malariae is the third most prevalent human malaria-causing species and has a patchy, but ample distribution in the world. Humans can host the parasite for years without presenting significant symptoms, turning its diagnosis and control into a difficult task. Here, we investigated the immunogenicity of recombinant proteins of P. malariae MSP1. Methods Five regions of PmMSP1 were expressed in Escherichia coli as GST-fusion proteins and immunized in BALB/c mice. The specificity, subtyping, and affinity of raised antibodies were evaluated by enzyme-linked immunosorbent assays. Cellular immune responses were analyzed by lymphoproliferation assays and cytokine levels produced by splenocytes were detected by cytometry. Results We found that N-terminal, central regions, and PmMSP119 are strongly immunogenic in mice. After three doses, the induced immune responses remained high for 70 days. While antibodies induced after immunization with N-terminal and central regions showed similar affinities to the target antigens, affinities of IgG against PmMSP119 were higher. All proteins induced similar antibody subclass patterns (predominantly IgG1, IgG2a, and IgG2b), characterizing a mixed Th1/Th2 response. Further, autologous stimulation of splenocytes from immunized mice led to the secretion of IL2 and IL4, independently of the antigen used. Importantly, IgG from P. malariae-exposed individuals reacted against PmMSP1 recombinant proteins with a high specificity. On the other hand, sera from P. vivax or P. falciparum-infected individuals did not react at all against recombinant PmMSP1 proteins. Conclusion Recombinant PmMSP1 proteins are very useful diagnostic markers of P. malariae in epidemiological studies or in the differential diagnosis of malaria caused by this species. Immunization with recombinant PmMSP1 proteins resulted in a significant humoral immune response, which may turn them potential component candidates for a vaccine against P. malariae.
Collapse
Affiliation(s)
- Yelina B. Elizardez
- Núcleo de Estudos em Malária, Superintendência de Controle de Endemias/Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil
| | - Wesley L. Fotoran
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Andrés J. Galisteo Junior
- Laboratório de Protozoologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil
| | - Izilda Curado
- Laboratório de Imunoepidemiologia, Superintendência de Controle de Endemias, São Paulo, Brazil
| | - Norival Kesper Junior
- Laboratório de Protozoologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil
| | - Eliana F. Monteiro
- Núcleo de Estudos em Malária, Superintendência de Controle de Endemias/Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil
| | - Irineu Romero Neto
- Laboratório de Protozoologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil
| | - Gerhard Wunderlich
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Karin Kirchgatter
- Núcleo de Estudos em Malária, Superintendência de Controle de Endemias/Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
16
|
Mohring F, Hart MN, Rawlinson TA, Henrici R, Charleston JA, Diez Benavente E, Patel A, Hall J, Almond N, Campino S, Clark TG, Sutherland CJ, Baker DA, Draper SJ, Moon RW. Rapid and iterative genome editing in the malaria parasite Plasmodium knowlesi provides new tools for P. vivax research. eLife 2019; 8:45829. [PMID: 31205002 PMCID: PMC6579517 DOI: 10.7554/elife.45829] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022] Open
Abstract
Tackling relapsing Plasmodium vivax and zoonotic Plasmodium knowlesi infections is critical to reducing malaria incidence and mortality worldwide. Understanding the biology of these important and related parasites was previously constrained by the lack of robust molecular and genetic approaches. Here, we establish CRISPR-Cas9 genome editing in a culture-adapted P. knowlesi strain and define parameters for optimal homology-driven repair. We establish a scalable protocol for the production of repair templates by PCR and demonstrate the flexibility of the system by tagging proteins with distinct cellular localisations. Using iterative rounds of genome-editing we generate a transgenic line expressing P. vivax Duffy binding protein (PvDBP), a lead vaccine candidate. We demonstrate that PvDBP plays no role in reticulocyte restriction but can alter the macaque/human host cell tropism of P. knowlesi. Critically, antibodies raised against the P. vivax antigen potently inhibit proliferation of this strain, providing an invaluable tool to support vaccine development.
Collapse
Affiliation(s)
- Franziska Mohring
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Melissa Natalie Hart
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - Ryan Henrici
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - James A Charleston
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ernest Diez Benavente
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Avnish Patel
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Joanna Hall
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control, Health Protection Agency, Hertfordshire, United Kingdom
| | - Neil Almond
- Division of Infectious Disease Diagnostics, National Institute for Biological Standards and Control, Health Protection Agency, Hertfordshire, United Kingdom
| | - Susana Campino
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Taane G Clark
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Colin J Sutherland
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - David A Baker
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Robert William Moon
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
17
|
Alanine DGW, Quinkert D, Kumarasingha R, Mehmood S, Donnellan FR, Minkah NK, Dadonaite B, Diouf A, Galaway F, Silk SE, Jamwal A, Marshall JM, Miura K, Foquet L, Elias SC, Labbé GM, Douglas AD, Jin J, Payne RO, Illingworth JJ, Pattinson DJ, Pulido D, Williams BG, de Jongh WA, Wright GJ, Kappe SHI, Robinson CV, Long CA, Crabb BS, Gilson PR, Higgins MK, Draper SJ. Human Antibodies that Slow Erythrocyte Invasion Potentiate Malaria-Neutralizing Antibodies. Cell 2019; 178:216-228.e21. [PMID: 31204103 PMCID: PMC6602525 DOI: 10.1016/j.cell.2019.05.025] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 03/05/2019] [Accepted: 05/13/2019] [Indexed: 12/30/2022]
Abstract
The Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) is the leading target for next-generation vaccines against the disease-causing blood-stage of malaria. However, little is known about how human antibodies confer functional immunity against this antigen. We isolated a panel of human monoclonal antibodies (mAbs) against PfRH5 from peripheral blood B cells from vaccinees in the first clinical trial of a PfRH5-based vaccine. We identified a subset of mAbs with neutralizing activity that bind to three distinct sites and another subset of mAbs that are non-functional, or even antagonistic to neutralizing antibodies. We also identify the epitope of a novel group of non-neutralizing antibodies that significantly reduce the speed of red blood cell invasion by the merozoite, thereby potentiating the effect of all neutralizing PfRH5 antibodies as well as synergizing with antibodies targeting other malaria invasion proteins. Our results provide a roadmap for structure-guided vaccine development to maximize antibody efficacy against blood-stage malaria.
Collapse
Affiliation(s)
- Daniel G W Alanine
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK; Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Doris Quinkert
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | | | - Shahid Mehmood
- Department of Chemistry, University of Oxford, Oxford OX1 3QZ, UK
| | - Francesca R Donnellan
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Nana K Minkah
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Ave. N., #500, Seattle, WA 98109, USA
| | - Bernadeta Dadonaite
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Francis Galaway
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - Sarah E Silk
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Abhishek Jamwal
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Jennifer M Marshall
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Lander Foquet
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Ave. N., #500, Seattle, WA 98109, USA
| | - Sean C Elias
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Geneviève M Labbé
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Alexander D Douglas
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Jing Jin
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Ruth O Payne
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Joseph J Illingworth
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - David J Pattinson
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - David Pulido
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Barnabas G Williams
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Willem A de Jongh
- ExpreS(2)ion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, Hørsholm 2970, Denmark
| | - Gavin J Wright
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Ave. N., #500, Seattle, WA 98109, USA
| | - Carol V Robinson
- Department of Chemistry, University of Oxford, Oxford OX1 3QZ, UK
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Brendan S Crabb
- Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Paul R Gilson
- Burnet Institute, 85 Commercial Road, Melbourne, VIC 3004, Australia
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK.
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| |
Collapse
|
18
|
Douglas AD, Baldeviano GC, Jin J, Miura K, Diouf A, Zenonos ZA, Ventocilla JA, Silk SE, Marshall JM, Alanine DGW, Wang C, Edwards NJ, Leiva KP, Gomez-Puerta LA, Lucas CM, Wright GJ, Long CA, Royal JM, Draper SJ. A defined mechanistic correlate of protection against Plasmodium falciparum malaria in non-human primates. Nat Commun 2019; 10:1953. [PMID: 31028254 PMCID: PMC6486575 DOI: 10.1038/s41467-019-09894-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/02/2019] [Indexed: 01/25/2023] Open
Abstract
Malaria vaccine design and prioritization has been hindered by the lack of a mechanistic correlate of protection. We previously demonstrated a strong association between protection and merozoite-neutralizing antibody responses following vaccination of non-human primates against Plasmodium falciparum reticulocyte binding protein homolog 5 (PfRH5). Here, we test the mechanism of protection. Using mutant human IgG1 Fc regions engineered not to engage complement or FcR-dependent effector mechanisms, we produce merozoite-neutralizing and non-neutralizing anti-PfRH5 chimeric monoclonal antibodies (mAbs) and perform a passive transfer-P. falciparum challenge study in Aotus nancymaae monkeys. At the highest dose tested, 6/6 animals given the neutralizing PfRH5-binding mAb c2AC7 survive the challenge without treatment, compared to 0/6 animals given non-neutralizing PfRH5-binding mAb c4BA7 and 0/6 animals given an isotype control mAb. Our results address the controversy regarding whether merozoite-neutralizing antibody can cause protection against P. falciparum blood-stage infections, and highlight the quantitative challenge of achieving such protection.
Collapse
Affiliation(s)
- Alexander D Douglas
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK.
| | - G Christian Baldeviano
- US Naval Medical Research Unit No. 6 (NAMRU-6), Av. Venezuela Cuadra 36, Bellavista, Callao, Peru
| | - Jing Jin
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Zenon A Zenonos
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Cambridge, CB10 1SA, UK
| | - Julio A Ventocilla
- US Naval Medical Research Unit No. 6 (NAMRU-6), Av. Venezuela Cuadra 36, Bellavista, Callao, Peru
| | - Sarah E Silk
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Jennifer M Marshall
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Daniel G W Alanine
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Chuan Wang
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Nick J Edwards
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Karina P Leiva
- US Naval Medical Research Unit No. 6 (NAMRU-6), Av. Venezuela Cuadra 36, Bellavista, Callao, Peru
| | - Luis A Gomez-Puerta
- US Naval Medical Research Unit No. 6 (NAMRU-6), Av. Venezuela Cuadra 36, Bellavista, Callao, Peru
| | - Carmen M Lucas
- US Naval Medical Research Unit No. 6 (NAMRU-6), Av. Venezuela Cuadra 36, Bellavista, Callao, Peru
| | - Gavin J Wright
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Cambridge, CB10 1SA, UK
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Joseph M Royal
- US Naval Medical Research Unit No. 6 (NAMRU-6), Av. Venezuela Cuadra 36, Bellavista, Callao, Peru
| | - Simon J Draper
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK.
| |
Collapse
|
19
|
Abstract
The development of highly effective and durable vaccines against the human malaria parasites Plasmodium falciparum and P. vivax remains a key priority. Decades of endeavor have taught that achieving this goal will be challenging; however, recent innovation in malaria vaccine research and a diverse pipeline of novel vaccine candidates for clinical assessment provides optimism. With first-generation pre-erythrocytic vaccines aiming for licensure in the coming years, it is important to reflect on how next-generation approaches can improve on their success. Here we review the latest vaccine approaches that seek to prevent malaria infection, disease, and transmission and highlight some of the major underlying immunological and molecular mechanisms of protection. The synthesis of rational antigen selection, immunogen design, and immunization strategies to induce quantitatively and qualitatively improved immune effector mechanisms offers promise for achieving sustained high-level protection.
Collapse
|
20
|
Siau A, Huang X, Loh HP, Zhang N, Meng W, Sze SK, Renia L, Preiser P. Immunomic Identification of Malaria Antigens Associated With Protection in Mice. Mol Cell Proteomics 2019; 18:837-853. [PMID: 30718293 DOI: 10.1074/mcp.ra118.000997] [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: 07/30/2018] [Revised: 01/22/2019] [Indexed: 11/06/2022] Open
Abstract
Efforts to develop vaccines against malaria represent a major research target. The observations that 1) sterile protection can be obtained when the host is exposed to live parasites and 2) the immunity against blood stage parasite is principally mediated by protective antibodies suggest that a protective vaccine is feasible. However, only a small number of proteins have been investigated so far and most of the Plasmodium proteome has yet to be explored. To date, only few immunodominant antigens have emerged for testing in clinical trials but no formulation has led to substantial protection in humans. The nature of parasite molecules associated with protection remains elusive. Here, immunomic screening of mice immune sera with different protection efficiencies against the whole parasite proteome allowed us to identify a large repertoire of antigens validated by screening a library expressing antigens. The calculation of weighted scores reflecting the likelihood of protection of each antigen using five predictive criteria derived from immunomic and proteomic data sets, highlighted a priority list of protective antigens. Altogether, the approach sheds light on conserved antigens across Plasmodium that are amenable to targeting by the host immune system upon merozoite invasion and blood stage development. Most of these antigens have preliminary protection data but have not been widely considered as candidate for vaccine trials, opening new perspectives that overcome the limited choice of immunodominant, poorly protective vaccines currently being the focus of malaria vaccine researches.
Collapse
Affiliation(s)
- Anthony Siau
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore;.
| | - Ximei Huang
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore;; From the ‡Nanyang Technological University, School of Biological Sciences, Singapore
| | - Han Ping Loh
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore;; From the ‡Nanyang Technological University, School of Biological Sciences, Singapore
| | - Neng Zhang
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore
| | - Wei Meng
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore
| | - Siu Kwan Sze
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore
| | - Laurent Renia
- §Singapore Immunology Network (SIgN), A*STAR, Biopolis, Singapore
| | - Peter Preiser
- From the ‡Nanyang Technological University, School of Biological Sciences, Singapore;.
| |
Collapse
|
21
|
Abstract
The blood stage of the malaria parasite life cycle is responsible for all the clinical symptoms of malaria. During the blood stage, Plasmodium merozoites invade and multiply within host red blood cells (RBCs). Here, we review the progress made, challenges faced, and new strategies available for the development of blood stage malaria vaccines. We discuss our current understanding of immune responses against blood stages and the status of clinical development of various blood stage malaria vaccine candidates. We then discuss possible paths forward to develop effective blood stage malaria vaccines. This includes a discussion of protective immune mechanisms that can be elicited to target blood stage parasites, novel delivery systems, immunoassays and animal models to optimize vaccine candidates in preclinical studies, and use of challenge models to get an early readout of vaccine efficacy.
Collapse
|
22
|
Thái TL, Jun H, Lee J, Kang JM, Lê HG, Lin K, Thant KZ, Sohn WM, Kim TS, Na BK. Genetic diversity of merozoite surface protein-1 C-terminal 42 kDa of Plasmodium falciparum (PfMSP-1 42) may be greater than previously known in global isolates. Parasit Vectors 2018; 11:455. [PMID: 30081943 PMCID: PMC6080494 DOI: 10.1186/s13071-018-3027-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/20/2018] [Indexed: 11/16/2022] Open
Abstract
Background The C-terminal 42 kDa region of merozoite surface protein-1 of Plasmodium falciparum (PfMSP-142) is the target of an immune response. It has been recognised as one of the promising candidate antigens for a blood-stage malaria vaccine. Genetic structure of PfMSP-142 has been considered to be largely conserved in the P. falciparum population. However, only limited information is currently available. This study aimed to analyse genetic diversity and the effect of natural selection on PfMSP-142 among the Myanmar P. falciparum population and compare them with publicly available PfMSP-142 from global P. falciparum populations. Methods A total of 69 P. falciparum clinical isolates collected from Myanmar malaria patients in Upper Myanmar in 2015 were used. The PfMSP-142 region was amplified by polymerase chain reaction, cloned and sequenced. Genetic structure and natural selection of this region were analysed using MEGA4 and DnaSP programs. Polymorphic nature and natural selection in global PfMSP-142 were also investigated. Results All three allele types (MAD20, K1, and RO33) of PfMSP-142 were identified in Myanmar isolates of P. falciparum. Myanmar PfMSP-142 displayed genetic diversity. Most polymorphisms were scattered in blocks 16 and 17. Polymorphisms observed in Myanmar PfMSP-142 showed a similar pattern to those of global PfMSP-142; however, they were not identical to each other. Genetic diversity of Myanmar PfMSP-142 was relatively lower than that of PfMSP-142 from different geographical regions. Evidence of natural selection and recombination were found. Comparative analysis of genetic polymorphism and natural selection in the global PfMSP-142 population suggested that this region was not tightly conserved in global PfMSP-142 as previously thought and is under the complicated influence of natural selection and recombination. Conclusions Global PfMSP-142 revealed limited, but non-negligible, genetic diversity by allele types and geographical origins. Complicated natural selection and potential recombination might have occurred in global PfMSP-142. Comprehensive monitoring of genetic diversity for global PfMSP-142 would be needed to better understand the polymorphic nature and evolutionary aspect of PfMSP-142 in the global P. falciparum population. More thought would be necessary for designing a vaccine based on PfMSP-142. Electronic supplementary material The online version of this article (10.1186/s13071-018-3027-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Thị Lam Thái
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Hojong Jun
- Department of Tropical Medicine, and Inha Research Institute for Medical Sciences, Inha University College of Medicine, Incheon, 22212, Republic of Korea
| | - Jinyoung Lee
- Department of Tropical Medicine, and Inha Research Institute for Medical Sciences, Inha University College of Medicine, Incheon, 22212, Republic of Korea
| | - Jung-Mi Kang
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Hương Giang Lê
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Khin Lin
- Department of Medical Research Pyin Oo Lwin Branch, Pyin Oo Lwin, Myanmar
| | - Kyaw Zin Thant
- Department of Medical Research, Ministry of Health and Sports, Yangon, Myanmar
| | - Woon-Mok Sohn
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea
| | - Tong-Soo Kim
- Department of Tropical Medicine, and Inha Research Institute for Medical Sciences, Inha University College of Medicine, Incheon, 22212, Republic of Korea.
| | - Byoung-Kuk Na
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea. .,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea.
| |
Collapse
|
23
|
Arévalo-Pinzón G, González-González M, Suárez CF, Curtidor H, Carabias-Sánchez J, Muro A, LaBaer J, Patarroyo MA, Fuentes M. Self-assembling functional programmable protein array for studying protein-protein interactions in malaria parasites. Malar J 2018; 17:270. [PMID: 30016987 PMCID: PMC6050706 DOI: 10.1186/s12936-018-2414-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 07/10/2018] [Indexed: 12/30/2022] Open
Abstract
Background Plasmodium vivax is the most widespread malarial species, causing significant morbidity worldwide. Knowledge is limited regarding the molecular mechanism of invasion due to the lack of a continuous in vitro culture system for these species. Since protein–protein and host–cell interactions play an essential role in the microorganism’s invasion and replication, elucidating protein function during invasion is critical when developing more effective control methods. Nucleic acid programmable protein array (NAPPA) has thus become a suitable technology for studying protein–protein and host–protein interactions since producing proteins through the in vitro transcription/translation (IVTT) method overcomes most of the drawbacks encountered to date, such as heterologous protein production, stability and purification. Results Twenty P. vivax proteins on merozoite surface or in secretory organelles were selected and successfully cloned using gateway technology. Most constructs were displayed in the array expressed in situ, using the IVTT method. The Pv12 protein was used as bait for evaluating array functionality and co-expressed with P. vivax cDNA display in the array. It was found that Pv12 interacted with Pv41 (as previously described), as well as PvMSP142kDa, PvRBP1a, PvMSP8 and PvRAP1. Conclusions NAPPA is a high-performance technique enabling co-expression of bait and query in situ, thereby enabling interactions to be analysed rapidly and reproducibly. It offers a fresh alternative for studying protein–protein and ligand–receptor interactions regarding a parasite which is difficult to cultivate (i.e. P. vivax). Electronic supplementary material The online version of this article (10.1186/s12936-018-2414-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gabriela Arévalo-Pinzón
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,PhD Programme in Biomedical and Biological Sciences, Universidad del Rosario, Carrera 24 # 63C-69, Bogotá, Colombia
| | - María González-González
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain.,Department of Medicine and General Cytometry Service-Nucleus, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain
| | - Carlos Fernando Suárez
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A.), Calle 222 # 55-37, Bogotá, Colombia
| | - Hernando Curtidor
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 # 63C-69, Bogotá, Colombia
| | | | - Antonio Muro
- Unidad de Investigación Enfermedades Infecciosas y Tropicales (e-INTRO), Instituto de Investigación Biomédica de Salamanca-Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (IBSAL-CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Universitario Miguel de Unamuno s/n, 37007, Salamanca, Spain
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Manuel Alfonso Patarroyo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 # 63C-69, Bogotá, Colombia
| | - Manuel Fuentes
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain. .,Department of Medicine and General Cytometry Service-Nucleus, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain.
| |
Collapse
|
24
|
Mehrizi AA, Rezvani N, Zakeri S, Gholami A, Babaeekhou L. Poly(I:C) adjuvant strongly enhances parasite-inhibitory antibodies and Th1 response against Plasmodium falciparum merozoite surface protein-1 (42-kDa fragment) in BALB/c mice. Med Microbiol Immunol 2018; 207:151-166. [PMID: 29397427 DOI: 10.1007/s00430-018-0535-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 01/10/2018] [Indexed: 10/18/2022]
Abstract
Malaria vaccine development has been confronted with various challenges such as poor immunogenicity of malaria vaccine candidate antigens, which is considered as the main challenge. However, this problem can be managed using appropriate formulations of antigens and adjuvants. Poly(I:C) is a potent Th1 inducer and a human compatible adjuvant capable of stimulating both B- and T-cell immunity. Plasmodium falciparum merozoite surface protein 142 (PfMSP-142) is a promising vaccine candidate for blood stage of malaria that has faced several difficulties in clinical trials, mainly due to improper adjuvants. Therefore, in the current study, poly(I:C), as a potent Th1 inducer adjuvant, was evaluated to improve the immunogenicity of recombinant PfMSP-142, when compared to CFA/IFA, as reference adjuvant. Poly(I:C) produced high level and titers of anti-PfMSP-142 IgG antibodies in which was comparable to CFA/IFA adjuvant. In addition, PfMSP-142 formulated with poly(I:C) elicited a higher ratio of IFN-γ/IL-4 (23.9) and IgG2a/IgG1 (3.77) with more persistent, higher avidity, and titer of IgG2a relative to CFA/IFA, indicating a potent Th1 immune response. Poly(I:C) could also help to induce anti-PfMSP-142 antibodies with higher growth-inhibitory activity than CFA/IFA. Altogether, the results of the current study demonstrated that poly(I:C) is a potent adjuvant that can be appropriate for being used in PfMSP-142-based vaccine formulations.
Collapse
Affiliation(s)
- Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran.
| | - Niloufar Rezvani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran.,Department of Biology, Islamshahr Branch, Islamic Azad University, Islamshahr, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Atefeh Gholami
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Laleh Babaeekhou
- Department of Biology, Islamshahr Branch, Islamic Azad University, Islamshahr, Tehran, Iran
| |
Collapse
|
25
|
Merozoite Surface Protein 1 from Plasmodium falciparum Is a Major Target of Opsonizing Antibodies in Individuals with Acquired Immunity against Malaria. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00155-17. [PMID: 28877929 DOI: 10.1128/cvi.00155-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/31/2017] [Indexed: 11/20/2022]
Abstract
Naturally acquired immunity against malaria is largely mediated by serum antibodies controlling levels of blood-stage parasites. A limited understanding of the antigenic targets and functional mechanisms of protective antibodies has hampered the development of efficient malaria vaccines. Besides directly inhibiting the growth of Plasmodium parasites, antibodies can opsonize merozoites and recruit immune effector cells such as monocytes and neutrophils. Antibodies against the vaccine candidate merozoite surface protein 1 (MSP-1) are acquired during natural infections and have been associated with protection against malaria in several epidemiological studies. Here we analyzed serum antibodies from semi-immune individuals from Burkina Faso for their potential (i) to directly inhibit the growth of P. falciparum blood stages in vitro and (ii) to opsonize merozoites and to induce the antibody-dependent respiratory burst (ADRB) activity of neutrophils. While a few sera that directly inhibited the growth of P. falciparum blood stages were identified, immunoglobulin G (IgG) from all individuals clearly mediated the activation of neutrophils. The level of neutrophil activation correlated with levels of antibodies to MSP-1, and affinity-purified MSP-1-specific antibodies elicited ADRB activity. Furthermore, immunization of nonhuman primates with recombinant full-size MSP-1 induced antibodies that efficiently opsonized P. falciparum merozoites. Reversing the function by preincubation with recombinant antigens allowed us to quantify the contribution of MSP-1 to the antiparasitic effect of serum antibodies. Our data suggest that MSP-1, especially the partially conserved subunit MSP-183, is a major target of opsonizing antibodies acquired during natural exposure to malaria. Induction of opsonizing antibodies might be a crucial effector mechanism for MSP-1-based malaria vaccines.
Collapse
|
26
|
Identification of Heparin Modifications and Polysaccharide Inhibitors of Plasmodium falciparum Merozoite Invasion That Have Potential for Novel Drug Development. Antimicrob Agents Chemother 2017; 61:AAC.00709-17. [PMID: 28893781 DOI: 10.1128/aac.00709-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/03/2017] [Indexed: 11/20/2022] Open
Abstract
Despite recent successful control efforts, malaria remains a leading global health burden. Alarmingly, resistance to current antimalarials is increasing and the development of new drug families is needed to maintain malaria control. Current antimalarials target the intraerythrocytic developmental stage of the Plasmodium falciparum life cycle. However, the invasive extracellular parasite form, the merozoite, is also an attractive target for drug development. We have previously demonstrated that heparin-like molecules, including those with low molecular weights and low anticoagulant activities, are potent and specific inhibitors of merozoite invasion and blood-stage replication. Here we tested a large panel of heparin-like molecules and sulfated polysaccharides together with various modified chemical forms for their inhibitory activity against P. falciparum merozoite invasion. We identified chemical modifications that improve inhibitory activity and identified several additional sulfated polysaccharides with strong inhibitory activity. These studies have important implications for the further development of heparin-like molecules as antimalarial drugs and for understanding merozoite invasion.
Collapse
|
27
|
Burel JG, Apte SH, Groves PL, Boyle MJ, Langer C, Beeson JG, McCarthy JS, Doolan DL. Dichotomous miR expression and immune responses following primary blood-stage malaria. JCI Insight 2017; 2:93434. [PMID: 28768914 DOI: 10.1172/jci.insight.93434] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/29/2017] [Indexed: 01/12/2023] Open
Abstract
Clinical responses to infection or vaccination and the development of effective immunity are characterized in humans by a marked interindividual variability. To gain an insight into the factors affecting this variability, we used a controlled human infection system to study early immune events following primary infection of healthy human volunteers with blood-stage Plasmodium falciparum malaria. By day 4 of infection, a dichotomous pattern of high or low expression of a defined set of microRNAs (miRs) emerged in volunteers that correlated with variation in parasite growth rate. Moreover, high-miR responders had higher numbers of activated CD4+ T cells, and developed significantly enhanced antimalarial antibody responses. Notably, a set of 17 miRs was identified in the whole blood of low-miR responders prior to infection that differentiated them from high-miR responders. These data implicate preexisting host factors as major determinants in the ability to effectively respond to primary malaria infection.
Collapse
Affiliation(s)
- Julie G Burel
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,The University of Queensland, Brisbane, Queensland, Australia
| | - Simon H Apte
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Penny L Groves
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Michelle J Boyle
- Menzies School of Health Research, Darwin, Northern Territory, Australia.,Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - Christine Langer
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - James G Beeson
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia.,Department of Microbiology, Monash University, Victoria, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,The University of Queensland, Brisbane, Queensland, Australia
| | - Denise L Doolan
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| |
Collapse
|
28
|
Srinivasan P, Baldeviano GC, Miura K, Diouf A, Ventocilla JA, Leiva KP, Lugo-Roman L, Lucas C, Orr-Gonzalez S, Zhu D, Villasante E, Soisson L, Narum DL, Pierce SK, Long CA, Diggs C, Duffy PE, Lescano AG, Miller LH. A malaria vaccine protects Aotus monkeys against virulent Plasmodium falciparum infection. NPJ Vaccines 2017; 2. [PMID: 28804644 PMCID: PMC5551459 DOI: 10.1038/s41541-017-0015-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The Plasmodium falciparum protein, apical membrane antigen 1 forms a complex with another parasite protein, rhoptry neck protein 2, to initiate junction formation with the erythrocyte and is essential for merozoite invasion during the blood stage of infection. Consequently, apical membrane antigen 1 has been a target of vaccine development but vaccination with apical membrane antigen 1 alone in controlled human malaria infections failed to protect and showed limited efficacy in field trials. Here we show that vaccination with AMA1–RON2L complex in Freund’s adjuvant protects Aotus monkeys against a virulent Plasmodium falciparum infection. Vaccination with AMA1 alone gave only partial protection, delaying infection in one of eight animals. However, the AMA1–RON2L complex vaccine completely protected four of eight monkeys and substantially delayed infection (>25 days) in three of the other four animals. Interestingly, antibodies from monkeys vaccinated with the AMA1–RON2L complex had significantly higher neutralizing activity than antibodies from monkeys vaccinated with AMA1 alone. Importantly, we show that antibodies from animals vaccinated with the complex have significantly higher neutralization activity against non-vaccine type parasites. We suggest that vaccination with the AMA1–RON2L complex induces functional antibodies that better recognize AMA1 as it appears complexed with RON2 during merozoite invasion. These data justify progression of this next generation AMA1 vaccine towards human trials. A vaccine targeting a protein complex that allows malaria-causing parasite to enter red blood cells has been produced. Malaria caused by the parasite Plasmodium falciparum is an oft-deadly infectious disease without an effective vaccine. A team of researchers at the National Institutes of Health led by Prakash Srinivasan, currently at the Johns Hopkins Malaria Research Institute, United States, demonstrated the efficacy of a vaccine candidate that works by priming a host’s immune system to a parasitic protein complex required to form a junction with red blood cells, allowing entry and proliferation of the pathogen. The group’s vaccine conferred more effective protection in monkeys than prior candidates that targeted only one component of the parasitic protein complex. This research warrants a closer look into how this candidate, and others targeting the protein complex, can be used to prevent malaria in humans.
Collapse
Affiliation(s)
- Prakash Srinivasan
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Karina P Leiva
- US Naval Medical Research Unit No. 6 (NAMRU-6), Callao, Peru
| | - Luis Lugo-Roman
- US Naval Medical Research Unit No. 6 (NAMRU-6), Callao, Peru
| | - Carmen Lucas
- US Naval Medical Research Unit No. 6 (NAMRU-6), Callao, Peru
| | - Sachy Orr-Gonzalez
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Daming Zhu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Eileen Villasante
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, MD, USA
| | - Lorraine Soisson
- Malaria Vaccine Development Program, U.S. Agency for International Development, Washington, DC, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Carter Diggs
- Malaria Vaccine Development Program, U.S. Agency for International Development, Washington, DC, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Louis H Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
29
|
A Plasmodium vivax Plasmid DNA- and Adenovirus-Vectored Malaria Vaccine Encoding Blood-Stage Antigens AMA1 and MSP1 42 in a Prime/Boost Heterologous Immunization Regimen Partially Protects Aotus Monkeys against Blood-Stage Challenge. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00539-16. [PMID: 28179404 DOI: 10.1128/cvi.00539-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/06/2017] [Indexed: 01/30/2023]
Abstract
Malaria is caused by parasites of the genus Plasmodium, which are transmitted to humans by the bites of Anopheles mosquitoes. After the elimination of Plasmodium falciparum, it is predicted that Plasmodium vivax will remain an important cause of morbidity and mortality outside Africa, stressing the importance of developing a vaccine against P. vivax malaria. In this study, we assessed the immunogenicity and protective efficacy of two P. vivax antigens, apical membrane antigen 1 (AMA1) and the 42-kDa C-terminal fragment of merozoite surface protein 1 (MSP142) in a plasmid recombinant DNA prime/adenoviral (Ad) vector boost regimen in Aotus monkeys. Groups of 4 to 5 monkeys were immunized with plasmid DNA alone, Ad alone, prime/boost regimens with each antigen, prime/boost regimens with both antigens, and empty vector controls and then subjected to blood-stage challenge. The heterologous immunization regimen with the antigen pair was more protective than either antigen alone or both antigens delivered with a single vaccine platform, on the basis of their ability to induce the longest prepatent period and the longest time to the peak level of parasitemia, the lowest peak and mean levels of parasitemia, the smallest area under the parasitemia curve, and the highest self-cure rate. Overall, prechallenge MSP142 antibody titers strongly correlated with a decreased parasite burden. Nevertheless, a significant proportion of immunized animals developed anemia. In conclusion, the P. vivax plasmid DNA/Ad serotype 5 vaccine encoding blood-stage parasite antigens AMA1 and MSP142 in a heterologous prime/boost immunization regimen provided significant protection against blood-stage challenge in Aotus monkeys, indicating the suitability of these antigens and this regimen for further development.
Collapse
|
30
|
Hodgson SH, Llewellyn D, Silk SE, Milne KH, Elias SC, Miura K, Kamuyu G, Juma EA, Magiri C, Muia A, Jin J, Spencer AJ, Longley RJ, Mercier T, Decosterd L, Long CA, Osier FH, Hoffman SL, Ogutu B, Hill AVS, Marsh K, Draper SJ. Changes in Serological Immunology Measures in UK and Kenyan Adults Post-controlled Human Malaria Infection. Front Microbiol 2016; 7:1604. [PMID: 27790201 PMCID: PMC5061779 DOI: 10.3389/fmicb.2016.01604] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/26/2016] [Indexed: 11/15/2022] Open
Abstract
Background: The timing of infection is closely determined in controlled human malaria infection (CHMI) studies, and as such they provide a unique opportunity to dissect changes in immunological responses before and after a single infection. The first Kenyan Challenge Study (KCS) (Pan African Clinical Trial Registry: PACTR20121100033272) was performed in 2013 with the aim of establishing the CHMI model in Kenya. This study used aseptic, cryopreserved, attenuated Plasmodium falciparum sporozoites administered by needle and syringe (PfSPZ Challenge) and was the first to evaluate parasite dynamics post-CHMI in individuals with varying degrees of prior exposure to malaria. Methods: We describe detailed serological and functional immunological responses pre- and post-CHMI for participants in the KCS and compare these with those from malaria-naïve UK volunteers who also underwent CHMI (VAC049) (ClinicalTrials.gov NCT01465048) using PfSPZ Challenge. We assessed antibody responses to three key blood-stage merozoite antigens [merozoite surface protein 1 (MSP1), apical membrane protein 1 (AMA1), and reticulocyte-binding protein homolog 5 (RH5)] and functional activity using two candidate measures of anti-merozoite immunity; the growth inhibition activity (GIA) assay and the antibody-dependent respiratory burst activity (ADRB) assay. Results:Clear serological differences were observed pre- and post-CHMI by ELISA between malaria-naïve UK volunteers in VAC049, and Kenyan volunteers who had prior malaria exposure. Antibodies to AMA1 and schizont extract correlated with parasite multiplication rate (PMR) post-CHMI in KCS. Serum from volunteer 110 in KCS, who demonstrated a dramatically reduced PMR in vivo, had no in vitro GIA prior to CHMI but the highest level of ADRB activity. A significant difference in ADRB activity was seen between KCS volunteers with minimal and definite prior exposure to malaria and significant increases were seen in ADRB activity post-CHMI in Kenyan volunteers. Quinine and atovaquone/proguanil, previously assumed to be removed by IgG purification, were identified as likely giving rise to aberrantly high in vitro GIA results. Conclusions: The ADRB activity assay is a promising functional assay that warrants further investigation as a measure of prior exposure to malaria and predictor of control of parasite growth. The CHMI model can be used to evaluate potential measures of naturally-acquired immunity to malaria.
Collapse
Affiliation(s)
| | | | - Sarah E Silk
- The Jenner Institute, University of Oxford Oxford, UK
| | | | - Sean C Elias
- The Jenner Institute, University of Oxford Oxford, UK
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIH-National Institute of Allergy and Infectious Diseases Rockville, MD, USA
| | - Gathoni Kamuyu
- Centre for Geographical Medical Research (Coast), Kenya Medical Research Institute-Wellcome Trust Kilifi, Kenya
| | - Elizabeth A Juma
- Centre for Clinical Research, Kenya Medical Research InstituteNairobi, Kenya; Centre for Research in Therapeutic Sciences, Strathmore UniversityNairobi, Kenya
| | - Charles Magiri
- Centre for Clinical Research, Kenya Medical Research Institute Nairobi, Kenya
| | - Alfred Muia
- Centre for Clinical Research, Kenya Medical Research Institute Nairobi, Kenya
| | - Jing Jin
- The Jenner Institute, University of Oxford Oxford, UK
| | | | | | - Thomas Mercier
- Division of Clinical Pharmacology, Hôpital Beaumont, Université de Lausanne Lausanne, Switzerland
| | - Laurent Decosterd
- Division of Clinical Pharmacology, Hôpital Beaumont, Université de Lausanne Lausanne, Switzerland
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIH-National Institute of Allergy and Infectious Diseases Rockville, MD, USA
| | - Faith H Osier
- Centre for Geographical Medical Research (Coast), Kenya Medical Research Institute-Wellcome Trust Kilifi, Kenya
| | | | - Bernhards Ogutu
- Centre for Clinical Research, Kenya Medical Research InstituteNairobi, Kenya; Centre for Research in Therapeutic Sciences, Strathmore UniversityNairobi, Kenya
| | | | - Kevin Marsh
- Centre for Geographical Medical Research (Coast), Kenya Medical Research Institute-Wellcome TrustKilifi, Kenya; Department of Tropical Medicine, University of OxfordOxford, UK
| | | |
Collapse
|
31
|
Chen Q, Liang W, Qian F, Qian B, Cao J, Zhang D, Xu Y, Tang L. Rice-produced MSP142ofPlasmodium falciparumelicits antibodies that inhibit parasite growth in vitro. Parasite Immunol 2016; 38:635-41. [DOI: 10.1111/pim.12352] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 08/01/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Q. Chen
- National Institute of Parasitic Diseases; Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Tropical Diseases; Key Laboratory of Parasite and Vector Biology; Ministry of Health; Shanghai China
| | - W. Liang
- State Key Laboratory of Hybrid Rice; School of Life Sciences and Biotechnology; Shanghai Jiao Tong University; Shanghai China
| | - F. Qian
- Department of Rheumatology and Immunology; Changzheng Hospital; Second Military Medical University; Shanghai China
| | - B. Qian
- State Key Laboratory of Hybrid Rice; School of Life Sciences and Biotechnology; Shanghai Jiao Tong University; Shanghai China
| | - J. Cao
- National Institute of Parasitic Diseases; Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Tropical Diseases; Key Laboratory of Parasite and Vector Biology; Ministry of Health; Shanghai China
| | - D. Zhang
- State Key Laboratory of Hybrid Rice; School of Life Sciences and Biotechnology; Shanghai Jiao Tong University; Shanghai China
| | - Y. Xu
- National Institute of Parasitic Diseases; Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Tropical Diseases; Key Laboratory of Parasite and Vector Biology; Ministry of Health; Shanghai China
| | - L. Tang
- National Institute of Parasitic Diseases; Chinese Center for Disease Control and Prevention; WHO Collaborating Centre for Tropical Diseases; Key Laboratory of Parasite and Vector Biology; Ministry of Health; Shanghai China
| |
Collapse
|
32
|
Bowman NM, Juliano JJ, Snider CJ, Kharabora O, Meshnick SR, Vulule J, John CC, Moormann AM. Longevity of Genotype-Specific Immune Responses to Plasmodium falciparum Merozoite Surface Protein 1 in Kenyan Children from Regions of Different Malaria Transmission Intensity. Am J Trop Med Hyg 2016; 95:580-7. [PMID: 27481054 DOI: 10.4269/ajtmh.15-0710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 06/02/2016] [Indexed: 01/06/2023] Open
Abstract
Naturally acquired immunity to Plasmodium falciparum presents a changing landscape as malaria control programs and vaccine initiatives are implemented. Determining which immunologic indicators remain surrogates of past infection, as opposed to mediators of protection, led us to compare stability of immune responses across regions with divergent malaria transmission intensities. A repeat cross-sectional study of Kenyan children from a malaria-holoendemic area and an epidemic-prone area was used to examine longitudinal antibody and interferon-gamma (IFN-γ) responses to the 3D7 and FVO variants of merozoite surface protein 1 (MSP1). Antibodies to MSP1 were common in both study populations and did not significantly wane over a 21-month time period. IFN-γ responses were less frequent and rapidly disappeared in children after a prolonged period of no malaria transmission. Antibody and IFN-γ responses rarely correlated with each other; however, MSP1-specific IFN-γ response correlated with lack of concurrent P. falciparum parasitemia of the same genotype, though only statistically significantly in the malaria-holoendemic region (odds ratio = 0.31, 95% confidence interval = 0.12-0.84). This study affirms that antimalarial antibodies are informative for evaluation of history of malaria exposure within individuals, whereas cell-mediated immunity, though short lived under natural exposure conditions, might provide an assessment of recent infection and protection from parasitemia.
Collapse
Affiliation(s)
- Natalie M Bowman
- Division of Infectious Diseases, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Jonathan J Juliano
- Division of Infectious Diseases, University of North Carolina, School of Medicine, Chapel Hill, North Carolina. Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, North Carolina.
| | - Cynthia J Snider
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Oksana Kharabora
- Division of Infectious Diseases, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Steven R Meshnick
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - John Vulule
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Chandy C John
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University, Indianapolis, Indiana
| | - Ann M Moormann
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
33
|
Hjerrild KA, Jin J, Wright KE, Brown RE, Marshall JM, Labbé GM, Silk SE, Cherry CJ, Clemmensen SB, Jørgensen T, Illingworth JJ, Alanine DGW, Milne KH, Ashfield R, de Jongh WA, Douglas AD, Higgins MK, Draper SJ. Production of full-length soluble Plasmodium falciparum RH5 protein vaccine using a Drosophila melanogaster Schneider 2 stable cell line system. Sci Rep 2016; 6:30357. [PMID: 27457156 PMCID: PMC4960544 DOI: 10.1038/srep30357] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/04/2016] [Indexed: 01/27/2023] Open
Abstract
The Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) has recently emerged as a leading candidate antigen against the blood-stage human malaria parasite. However it has proved challenging to identify a heterologous expression platform that can produce a soluble protein-based vaccine in a manner compliant with current Good Manufacturing Practice (cGMP). Here we report the production of full-length PfRH5 protein using a cGMP-compliant platform called ExpreS(2), based on a Drosophila melanogaster Schneider 2 (S2) stable cell line system. Five sequence variants of PfRH5 were expressed that differed in terms of mutagenesis strategies to remove potential N-linked glycans. All variants bound the PfRH5 receptor basigin and were recognized by a panel of monoclonal antibodies. Analysis following immunization of rabbits identified quantitative and qualitative differences in terms of the functional IgG antibody response against the P. falciparum parasite. The antibodies induced by one protein variant were shown to be qualitatively similar to responses induced by other vaccine platforms. This work identifies Drosophila S2 cells as a clinically-relevant platform suited for the production of 'difficult-to-make' proteins from Plasmodium parasites, and identifies a PfRH5 sequence variant that can be used for clinical production of a non-glycosylated, soluble full-length protein vaccine immunogen.
Collapse
Affiliation(s)
- Kathryn A Hjerrild
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Jing Jin
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Katherine E Wright
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Rebecca E Brown
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Jennifer M Marshall
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Geneviève M Labbé
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Sarah E Silk
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Catherine J Cherry
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Stine B Clemmensen
- ExpreS2ion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, Hørsholm DK-2970, Denmark
| | - Thomas Jørgensen
- ExpreS2ion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, Hørsholm DK-2970, Denmark
| | - Joseph J Illingworth
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Daniel G W Alanine
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Kathryn H Milne
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Rebecca Ashfield
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Willem A de Jongh
- ExpreS2ion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, Hørsholm DK-2970, Denmark
| | - Alexander D Douglas
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| |
Collapse
|
34
|
Plasmodium falciparum infection and age influence parasite growth inhibition mediated by IgG in Beninese infants. Acta Trop 2016; 159:111-9. [PMID: 27001144 DOI: 10.1016/j.actatropica.2016.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/10/2016] [Accepted: 03/17/2016] [Indexed: 11/22/2022]
Abstract
Antibodies that impede the invasion of Plasmodium falciparum (P. falciparum) merozoites into erythrocytes play a critical role in anti-malarial immunity. The Growth Inhibition Assay (GIA) is an in vitro measure of the functional capacity of such antibodies to limit erythrocyte invasion and/or parasite growth. Up to now, it is unclear whether growth-inhibitory activity correlates with protection from clinical disease and there are inconsistent results from studies performed with GIA. Studies that have focused on the relationship between IgGs and their in vitro parasite Growth Inhibition Activity (GIAc) in infants aged less than two years old are rare. Here, we used clinical and parasitological data to precisely define symptomatic or asymptomatic infection with P. falciparum in groups of infants followed-up actively for 18 months post-natally. We quantified the levels of IgG1 and IgG3 directed to a panel of candidate P. falciparum vaccine antigens (AMA-1, MSP1, 2, 3 and GLURP) using ELISA and the functional activity of IgG was quantified using GIA. Data were then correlated with individuals' infection status. At 18 months of age, infants harbouring infections at the time of blood sampling had an average 19% less GIAc than those not infected (p=0.004, multivariate linear regression). GIAc decreased from 12 to 18 months of age (p=0.003, Wilcoxon matched pairs test). Antibody levels quantified at 18 months in infants were strongly correlated with their exposure to malarial infection, however GIAc was not correlated with malaria infectious status (asymptomatic and symptomatic groups). In conclusion, both infection status at blood draw and age influence parasite growth inhibition mediated by IgG in the GIA. Both factors must be taken into account when correlations between GIAc and anti-malarial protection or vaccine efficacy have to be made.
Collapse
|
35
|
Persistence and immunogenicity of chemically attenuated blood stage Plasmodium falciparum in Aotus monkeys. Int J Parasitol 2016; 46:581-91. [PMID: 27238088 DOI: 10.1016/j.ijpara.2016.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 04/05/2016] [Accepted: 05/03/2016] [Indexed: 12/24/2022]
Abstract
Malaria is a disease caused by a protozoan of the Plasmodium genus and results in 0.5-0.7million deaths per year. Increasing drug resistance of the parasite and insecticide resistance of mosquitoes necessitate alternative control measures. Numerous vaccine candidates have been identified but none have been able to induce robust, long-lived protection when evaluated in malaria endemic regions. Rodent studies have demonstrated that chemically attenuated blood stage parasites can persist at sub-patent levels and induce homologous and heterologous protection against malaria. Parasite-specific cellular responses were detected, with protection dependent on CD4+ T cells. To investigate this vaccine approach for Plasmodium falciparum, we characterised the persistence and immunogenicity of chemically attenuated P. falciparum FVO strain parasites (CAPs) in non-splenectomised Aotus nancymaae monkeys following administration of a single dose. Control monkeys received either normal red blood cells or wild-type parasites followed by drug treatment. Chemical attenuation was performed using tafuramycin A, which irreversibly binds to DNA. CAPs were detected in the peripheral blood for up to 2days following inoculation as determined by thick blood smears, and for up to 8days as determined by quantitative PCR. Parasite-specific IgG was not detected in monkeys that received CAPs; however, in vitro parasite-specific T cell proliferation was observed. Following challenge, the CAP monkeys developed an infection; however, one CAP monkey and the infection and drug-cure monkeys showed partial or complete resistance. These experiments lay the groundwork for further assessment of CAPs as a potential vaccine against malaria.
Collapse
|
36
|
Biryukov S, Angov E, Landmesser ME, Spring MD, Ockenhouse CF, Stoute JA. Complement and Antibody-mediated Enhancement of Red Blood Cell Invasion and Growth of Malaria Parasites. EBioMedicine 2016; 9:207-216. [PMID: 27333049 PMCID: PMC4972486 DOI: 10.1016/j.ebiom.2016.05.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/03/2016] [Accepted: 05/12/2016] [Indexed: 11/02/2022] Open
Abstract
Plasmodium falciparum malaria is a deadly pathogen. The invasion of red blood cells (RBCs) by merozoites is a target for vaccine development. Although anti-merozoite antibodies can block invasion in vitro, there is no efficacy in vivo. To explain this discrepancy we hypothesized that complement activation could enhance RBC invasion by binding to the complement receptor 1 (CR1). Here we show that a monoclonal antibody directed against the merozoite and human polyclonal IgG from merozoite vaccine recipients enhanced RBC invasion in a complement-dependent manner and that soluble CR1 inhibited this enhancement. Sialic acid-independent strains, that presumably are able to bind to CR1 via a native ligand, showed less complement-dependent enhancement of RBC invasion than sialic acid-dependent strains that do not utilize native CR1 ligands. Confocal fluorescent microscopy revealed that complement-dependent invasion resulted in aggregation of CR1 at the RBC surface in contact with the merozoite. Finally, total anti-P. berghei IgG enhanced parasite growth and C3 deficiency decreased parasite growth in mice. These results demonstrate, contrary to current views, that complement activation in conjunction with antibodies can paradoxically aid parasites invade RBCs and should be considered in future design and testing of merozoite vaccines.
Collapse
Affiliation(s)
- Sergei Biryukov
- Department of Microbiology and Immunology, Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Evelina Angov
- Walter Reed Army Institute of Research, Division of Malaria Vaccine Development, Silver Spring, MD 20910, United States
| | - Mary E Landmesser
- Department of Microbiology and Immunology, Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, PA 17033, United States; Department of Medicine, Division of Infectious Diseases and Epidemiology, Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, PA 17033, United States
| | - Michele D Spring
- Walter Reed Army Institute of Research, Division of Malaria Vaccine Development, Silver Spring, MD 20910, United States
| | | | - José A Stoute
- Department of Microbiology and Immunology, Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, PA 17033, United States; Department of Medicine, Division of Infectious Diseases and Epidemiology, Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, PA 17033, United States.
| |
Collapse
|
37
|
Ord RL, Rodriguez M, Lobo CA. Malaria invasion ligand RH5 and its prime candidacy in blood-stage malaria vaccine design. Hum Vaccin Immunother 2016; 11:1465-73. [PMID: 25844685 DOI: 10.1080/21645515.2015.1026496] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
With drug resistance to available therapeutics continuing to develop against Plasmodium falciparum malaria, the development of an effective vaccine candidate remains a major research goal. Successful interruption of invasion of parasites into erythrocytes during the blood stage of infection will prevent the severe clinical symptoms and complications associated with malaria. Previously studied blood stage antigens have highlighted the hurdles that are inherent to this life-cycle stage, namely that highly immunogenic antigens are also globally diverse, resulting in protection only against the vaccine strain, or that naturally acquired immunity to blood stage antigens do not always correlate with actual protection. The blood stage antigen reticulocyte binding homolog RH5 is essential for parasite viability, has globally limited diversity, and is associated with protection from disease. Here we summarize available information on this invasion ligand and recent findings that highlight its candidacy for inclusion in a blood-stage malaria vaccine.
Collapse
Affiliation(s)
- Rosalynn L Ord
- a Blood-Borne Parasites; Lindsley Kimball Research Institute; New York Blood Center ; New York , NY , USA
| | | | | |
Collapse
|
38
|
Jones DS, Rowe CG, Chen B, Reiter K, Rausch KM, Narum DL, Wu Y, Duffy PE. A Method for Producing Protein Nanoparticles with Applications in Vaccines. PLoS One 2016; 11:e0138761. [PMID: 26950441 PMCID: PMC4780713 DOI: 10.1371/journal.pone.0138761] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 09/03/2015] [Indexed: 11/18/2022] Open
Abstract
A practical method is described for synthesizing conjugated protein nanoparticles using thioether (thiol-maleimide) cross-linking chemistry. This method fills the need for a reliable and reproducible synthesis of protein conjugate vaccines for preclinical studies, which can be adapted to produce comparable material for clinical studies. The described method appears to be generally applicable to the production of nanoparticles from a variety of soluble proteins having different structural features. Examples presented include single-component particles of the malarial antigens AMA1, CSP and Pfs25, and two component particles comprised of those antigens covalently cross-linked with the immunogenic carrier protein EPA (a detoxified form of exotoxin A from Pseudomonas aeruginosa). The average molar masses (Mw) of particles in the different preparations ranged from 487 kDa to 3,420 kDa, with hydrodynamic radii (Rh) ranging from 12.1 nm to 38.3 nm. The antigenic properties and secondary structures of the proteins within the particles appear to be largely intact, with no significant changes seen in their far UV circular dichroism spectra, or in their ability to bind conformation-dependent monoclonal antibodies. Mice vaccinated with mixed particles of Pfs25 or CSP and EPA generated significantly greater antigen-specific antibody levels compared with mice vaccinated with the respective unmodified monomeric antigens, validating the potential of antigen-EPA nanoparticles as vaccines.
Collapse
Affiliation(s)
- David S. Jones
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
- * E-mail:
| | - Christopher G. Rowe
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - Beth Chen
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - Karine Reiter
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - Kelly M. Rausch
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - David L. Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - Yimin Wu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, Maryland, 20852, United States of America
| |
Collapse
|
39
|
Abstract
There have been significant decreases in malaria mortality and morbidity in the last 10-15 years, and the most advanced pre-erythrocytic malaria vaccine, RTS,S, received a positive opinion from European regulators in July 2015. However, no blood-stage vaccine has reached a phase III trial. The first part of this review summarizes the pros and cons of various assays and models that have been and will be used to predict the efficacy of blood-stage vaccines. In the second part, blood-stage vaccine candidates that showed some efficacy in human clinical trials or controlled human malaria infection models are discussed. Then, candidates under clinical investigation are described in the third part, and other novel candidates and strategies are reviewed in the last part.
Collapse
Affiliation(s)
- Kazutoyo Miura
- a Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Rockville , MD , USA
| |
Collapse
|
40
|
Cheong FW, Fong MY, Lau YL. Identification and characterization of epitopes on Plasmodium knowlesi merozoite surface protein-142 (MSP-142) using synthetic peptide library and phage display library. Acta Trop 2016; 154:89-94. [PMID: 26624919 DOI: 10.1016/j.actatropica.2015.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/07/2015] [Accepted: 11/09/2015] [Indexed: 10/22/2022]
Abstract
Plasmodium knowlesi can cause potentially life threatening human malaria. The Plasmodium merozoite surface protein-142 (MSP-142) is a potential target for malaria blood stage vaccine, and for diagnosis of malaria. Two epitope mapping techniques were used to identify the potential epitopes within P. knowlesi MSP-142. Nine and 14 potential epitopes were identified using overlapping synthetic peptide library and phage display library, respectively. Two regions on P. knowlesi MSP-142 (amino acid residues 37-95 and residues 240-289) were identified to be the potential dominant epitope regions. Two of the prominent epitopes, P10 (TAKDGMEYYNKMGELYKQ) and P31 (RCLLGFKEVGGKCVPASI), were evaluated using mouse model. P10- and P31-immunized mouse sera reacted with recombinant P. knowlesi MSP-142, with the IgG isotype distribution of IgG2b>IgG1>IgG2a>IgG3. Significant higher level of cytokines interferon-gamma and interleukin-2 was detected in P31-immunized mice. Both P10 and P31 could be the suitable epitope candidates to be used in malaria vaccine designs and immunodiagnostic assays, provided further evaluation is needed to validate the potential uses of these epitopes.
Collapse
|
41
|
Abstract
Malaria is one of the most serious infectious diseases with most of the severe disease
caused by Plasmodium falciparum (Pf). Naturally acquired immunity
develops over time after repeated infections and the development of antimalarial
antibodies is thought to play a crucial role. Neonates and young infants are relatively
protected from symptomatic malaria through mechanisms that are poorly understood. The
prevailing paradigm is that maternal antimalarial antibodies transferred to the fetus in
the last trimester of pregnancy protect the infant from early infections. These
antimalarial antibodies wane by approximately 6 months of age leaving the infant
vulnerable to malaria, however direct evidence supporting this epidemiologically based
paradigm is lacking. As infants are the target population for future malaria vaccines,
understanding how they begin to develop immunity to malaria and the gaps in their
responses is key. This review summarizes the antimalarial antibody responses detected in
infants and how they change over time. We focus primarily on Pf antibody responses and
will briefly mention Plasmodium vivax responses in infants.
Collapse
|
42
|
Abstract
With increasing malaria control and goals of malaria elimination, many endemic areas are transitioning from high-to-low-to-no malaria transmission. Reductions in transmission will impact on the development of naturally acquired immunity to malaria, which develops after repeated exposure to Plasmodium spp. However, it is currently unclear how declining transmission and malaria exposure will affect the development and maintenance of naturally acquired immunity. Here we review the key processes which underpin this knowledge; the amount of Plasmodium spp. exposure required to generate effective immune responses, the longevity of antibody responses and the ability to mount an effective response upon re-exposure through memory responses. Lastly we identify research priorities which will increase our understanding of how changing transmission will impact on malarial immunity.
Collapse
|
43
|
Contrasting Patterns of Serologic and Functional Antibody Dynamics to Plasmodium falciparum Antigens in a Kenyan Birth Cohort. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 23:104-16. [PMID: 26656119 PMCID: PMC4744923 DOI: 10.1128/cvi.00452-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/20/2015] [Indexed: 11/20/2022]
Abstract
IgG antibodies to Plasmodium falciparum are transferred from the maternal to fetal circulation during pregnancy, wane after birth, and are subsequently acquired in response to natural infection. We examined the dynamics of malaria antibody responses of 84 Kenyan infants from birth to 36 months of age by (i) serology, (ii) variant surface antigen (VSA) assay, (iii) growth inhibitory activity (GIA), and (iv) invasion inhibition assays (IIA) specific for merozoite surface protein 1 (MSP1) and sialic acid-dependent invasion pathway. Maternal antibodies in each of these four categories were detected in cord blood and decreased to their lowest level by approximately 6 months of age. Serologic antibodies to 3 preerythrocytic and 10 blood-stage antigens subsequently increased, reaching peak prevalence by 36 months. In contrast, antibodies measured by VSA, GIA, and IIA remained low even up to 36 months. Infants sensitized to P. falciparum in utero, defined by cord blood lymphocyte recall responses to malaria antigens, acquired antimalarial antibodies at the same rate as those who were not sensitized in utero, indicating that fetal exposure to malaria antigens did not affect subsequent infant antimalarial responses. Infants with detectable serologic antibodies at 12 months of age had an increased risk of P. falciparum infection during the subsequent 24 months. We conclude that serologic measures of antimalarial antibodies in children 36 months of age or younger represent biomarkers of malaria exposure rather than protection and that functional antibodies develop after 36 months of age in this population.
Collapse
|
44
|
Khan F, Porter M, Schwenk R, DeBot M, Saudan P, Dutta S. Head-to-Head Comparison of Soluble vs. Qβ VLP Circumsporozoite Protein Vaccines Reveals Selective Enhancement of NANP Repeat Responses. PLoS One 2015; 10:e0142035. [PMID: 26571021 PMCID: PMC4646581 DOI: 10.1371/journal.pone.0142035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/17/2015] [Indexed: 11/23/2022] Open
Abstract
Circumsporozoite protein (CSP) of Plasmodium falciparum is a promising malaria vaccine target. RTS,S, the most advanced malaria vaccine candidate consists of the central NANP repeat and carboxy-terminal region of CSP displayed on a hepatitis B virus-like particle (VLP). To build upon the success of RTS,S, we produced a near full-length Plasmodium falciparum CSP that also includes the conserved amino-terminal region of CSP. We recently showed that this soluble CSP, combined with a synthetic Toll-like-receptor-4 (TLR4) agonist in stable oil-in-water emulsion (GLA/SE), induces a potent and protective immune response in mice against transgenic parasite challenge. Here we have investigated whether the immunogenicity of soluble CSP could be further augmented by presentation on a VLP. Bacteriophage Qβ VLPs can be readily produced in E.coli, they have a diameter of 25 nm and contain packaged E. coli RNA which serves as a built in adjuvant through the activation of TLR7/8. CSP was chemically conjugated to Qβ and the CSP-Qβ vaccine immunogenicity and efficacy were compared to adjuvanted soluble CSP in the C57Bl/6 mouse model. When formulated with adjuvants lacking a TLR4 agonist (Alum, SE and Montanide) the Qβ-CSP induced higher anti-NANP repeat titers, higher levels of cytophilic IgG2b/c antibodies and a trend towards higher protection against transgenic parasite challenge as compared to soluble CSP formulated in the same adjuvant. The VLP and soluble CSP immunogenicity difference was most pronounced at low antigen dose, and within the CSP molecule, the titers against the NANP repeats were preferentially enhanced by Qβ presentation. While a TLR4 agonist enhanced the immunogenicity of soluble CSP to levels comparable to the VLP vaccine, the TLR4 agonist did not further improve the immunogenicity of the Qβ-CSP vaccine. The data presented here pave the way for further improvement in the Qβ conjugation chemistry and evaluation of both the Qβ-CSP and soluble CSP vaccines in the non-human primate model.
Collapse
Affiliation(s)
- Farhat Khan
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Mike Porter
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Robert Schwenk
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Margot DeBot
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Philippe Saudan
- Cytos Biotechnology, Wagistrasse 25, 8952 Schlieren, Switzerland
| | - Sheetij Dutta
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
- * E-mail:
| |
Collapse
|
45
|
Joos C, Varela ML, Mbengue B, Mansourou A, Marrama L, Sokhna C, Tall A, Trape JF, Touré A, Mercereau-Puijalon O, Perraut R. Antibodies to Plasmodium falciparum merozoite surface protein-1p19 malaria vaccine candidate induce antibody-dependent respiratory burst in human neutrophils. Malar J 2015; 14:409. [PMID: 26471813 PMCID: PMC4608189 DOI: 10.1186/s12936-015-0935-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/27/2015] [Indexed: 11/29/2022] Open
Abstract
Background Identification of plasmodial antigens targeted by protective immune mechanisms is important for malaria vaccine development. Among functional assays, the neutrophil antibody-dependent respiratory burst (ADRB) induced by opsonized Plasmodium falciparum merozoites has been correlated with acquired immunity to clinical malaria in endemic areas, but the target merozoite antigens are unknown. Here, the contribution of antibodies to the conserved C-terminal domain of the P. falciparum merozoite surface protein-1 (PfMSP1p19) in mediating ADRB was investigated in sera from individuals living in two Senegalese villages with differing malaria endemicity. Methods Anti-PfMSP1p19 antibody levels in sera from 233 villagers were investigated and the involvement of anti-PfMSP1p19 antibodies in ADRB was explored in a subset of samples using (1) isogenic P. falciparum parasite clones expressing P. falciparum or Plasmodium chabaudi MSP1p19; (2) PfMSP1p19-coated plaque ADRB; and, (3) ADRB triggering using sera depleted from PfMSP1p19 antibodies by absorption onto the baculovirus recombinant antigen. Results ADRB activity correlated with anti-PfMSP1p19 IgG levels (P < 10−3). A substantial contribution of PfMSP1p19 antibody responses to ADRB was confirmed (P < 10−4) in an age-adjusted linear regression model. PfMSP1p19 antibodies accounted for 33.1 % (range 7–54 %) and 33.2 % (range 0–70 %) of ADRB activity evaluated using isogenic merozoites (P < 10−3) and depleted sera (P = 0.0017), respectively. Coating of PfMSP1p19 on plates induced strong ADRB in anti-PfMSP1p19-positive sera. Conclusion These data show that naturally acquired P. falciparum MSP1p19 antibodies are potent inducers of neutrophil ADRB and support the development of PfMSP1p19-based malaria vaccine using ADRB assay as a functional surrogate for protection. Electronic supplementary material The online version of this article (doi:10.1186/s12936-015-0935-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charlotte Joos
- Unité d'Immunologie, Institut Pasteur de Dakar, Dakar, Senegal. .,Unité d'Immunologie Moléculaire des Parasites, Institut Pasteur, Paris, France.
| | | | - Babacar Mbengue
- Unité d'Immunogénétique/UCAD, Institut Pasteur de Dakar, Dakar, Senegal.
| | | | - Laurence Marrama
- Unité d'Epidémiologie, Institut Pasteur de Dakar, Dakar, Senegal.
| | - Cheikh Sokhna
- Institut de Recherche pour le Développement (IRD), URMITE, Dakar, Senegal.
| | - Adama Tall
- Unité d'Epidémiologie, Institut Pasteur de Dakar, Dakar, Senegal.
| | | | - Aissatou Touré
- Unité d'Immunologie, Institut Pasteur de Dakar, Dakar, Senegal.
| | | | - Ronald Perraut
- Unité d'Immunologie, Institut Pasteur de Dakar, Dakar, Senegal.
| |
Collapse
|
46
|
Douglas AD, Baldeviano GC, Lucas CM, Lugo-Roman LA, Crosnier C, Bartholdson SJ, Diouf A, Miura K, Lambert LE, Ventocilla JA, Leiva KP, Milne KH, Illingworth JJ, Spencer AJ, Hjerrild KA, Alanine DGW, Turner AV, Moorhead JT, Edgel KA, Wu Y, Long CA, Wright GJ, Lescano AG, Draper SJ. A PfRH5-based vaccine is efficacious against heterologous strain blood-stage Plasmodium falciparum infection in aotus monkeys. Cell Host Microbe 2015; 17:130-9. [PMID: 25590760 PMCID: PMC4297294 DOI: 10.1016/j.chom.2014.11.017] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 09/11/2014] [Accepted: 11/13/2014] [Indexed: 12/22/2022]
Abstract
Antigenic diversity has posed a critical barrier to vaccine development against the pathogenic blood-stage infection of the human malaria parasite Plasmodium falciparum. To date, only strain-specific protection has been reported by trials of such vaccines in nonhuman primates. We recently showed that P. falciparum reticulocyte binding protein homolog 5 (PfRH5), a merozoite adhesin required for erythrocyte invasion, is highly susceptible to vaccine-inducible strain-transcending parasite-neutralizing antibody. In vivo efficacy of PfRH5-based vaccines has not previously been evaluated. Here, we demonstrate that PfRH5-based vaccines can protect Aotus monkeys against a virulent vaccine-heterologous P. falciparum challenge and show that such protection can be achieved by a human-compatible vaccine formulation. Protection was associated with anti-PfRH5 antibody concentration and in vitro parasite-neutralizing activity, supporting the use of this in vitro assay to predict the in vivo efficacy of future vaccine candidates. These data suggest that PfRH5-based vaccines have potential to achieve strain-transcending efficacy in humans. Vaccines based on the P. falciparum merozoite antigen PfRH5 were tested in Aotus monkeys PfRH5-based vaccines afforded protection against heterologous strains of P. falciparum Protection correlated with anti-PfRH5 IgG concentration and in vivo neutralization
Collapse
Affiliation(s)
| | | | - Carmen M Lucas
- US Naval Medical Research Unit No. 6 (NAMRU-6), Lima, Peru
| | | | | | | | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Lynn E Lambert
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Rockville, MD 20852, USA
| | | | - Karina P Leiva
- US Naval Medical Research Unit No. 6 (NAMRU-6), Lima, Peru
| | | | | | | | | | | | | | | | | | - Yimin Wu
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Rockville, MD 20852, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | | | | | - Simon J Draper
- Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
47
|
Barry AE, Arnott A. Strategies for designing and monitoring malaria vaccines targeting diverse antigens. Front Immunol 2014; 5:359. [PMID: 25120545 PMCID: PMC4112938 DOI: 10.3389/fimmu.2014.00359] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/13/2014] [Indexed: 01/28/2023] Open
Abstract
After more than 50 years of intensive research and development, only one malaria vaccine candidate, “RTS,S,” has progressed to Phase 3 clinical trials. Despite only partial efficacy, this candidate is now forecast to become the first licensed malaria vaccine. Hence, more efficacious second-generation malaria vaccines that can significantly reduce transmission are urgently needed. This review will focus on a major obstacle hindering development of effective malaria vaccines: parasite antigenic diversity. Despite extensive genetic diversity in leading candidate antigens, vaccines have been and continue to be formulated using recombinant antigens representing only one or two strains. These vaccine strains represent only a small fraction of the diversity circulating in natural parasite populations, leading to escape of non-vaccine strains and challenging investigators’ abilities to measure strain-specific efficacy in vaccine trials. Novel strategies are needed to overcome antigenic diversity in order for vaccine development to succeed. Many studies have now cataloged the global diversity of leading Plasmodium falciparum and Plasmodium vivax vaccine antigens. In this review, we describe how population genetic approaches can be applied to this rich data source to predict the alleles that best represent antigenic diversity, polymorphisms that contribute to it, and to identify key polymorphisms associated with antigenic escape. We also suggest an approach to summarize the known global diversity of a given antigen to predict antigenic diversity, how to select variants that best represent the strains circulating in natural parasite populations and how to investigate the strain-specific efficacy of vaccine trials. Use of these strategies in the design and monitoring of vaccine trials will not only shed light on the contribution of genetic diversity to the antigenic diversity of malaria, but will also maximize the potential of future malaria vaccine candidates.
Collapse
Affiliation(s)
- Alyssa E Barry
- Division of Infection and Immunity, Walter and Eliza Hall Institute of Medical Research , Parkville, VIC , Australia ; Department of Medical Biology, The University of Melbourne , Parkville, VIC , Australia
| | - Alicia Arnott
- Division of Infection and Immunity, Walter and Eliza Hall Institute of Medical Research , Parkville, VIC , Australia ; Department of Medical Biology, The University of Melbourne , Parkville, VIC , Australia
| |
Collapse
|
48
|
Cavanagh DR, Kocken CHM, White JH, Cowan GJM, Samuel K, Dubbeld MA, der Wel AVV, Thomas AW, McBride JS, Arnot DE. Antibody responses to a novel Plasmodium falciparum merozoite surface protein vaccine correlate with protection against experimental malaria infection in Aotus monkeys. PLoS One 2014; 9:e83704. [PMID: 24421900 PMCID: PMC3885447 DOI: 10.1371/journal.pone.0083704] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 11/06/2013] [Indexed: 11/29/2022] Open
Abstract
The Block 2 region of the merozoite surface protein-1 (MSP-1) of Plasmodium falciparum has been identified as a target of protective immunity by a combination of seroepidemiology and parasite population genetics. Immunogenicity studies in small animals and Aotus monkeys were used to determine the efficacy of recombinant antigens derived from this region of MSP-1 as a potential vaccine antigen. Aotus lemurinus griseimembra monkeys were immunized three times with a recombinant antigen derived from the Block 2 region of MSP-1 of the monkey-adapted challenge strain, FVO of Plasmodium falciparum, using an adjuvant suitable for use in humans. Immunofluorescent antibody assays (IFA) against erythrocytes infected with P. falciparum using sera from the immunized monkeys showed that the MSP-1 Block 2 antigen induced significant antibody responses to whole malaria parasites. MSP-1 Block 2 antigen-specific enzyme-linked immunosorbent assays (ELISA) showed no significant differences in antibody titers between immunized animals. Immunized animals were challenged with the virulent P. falciparum FVO isolate and monitored for 21 days. Two out of four immunized animals were able to control their parasitaemia during the follow-up period, whereas two out of two controls developed fulminating parasitemia. Parasite-specific serum antibody titers measured by IFA were four-fold higher in protected animals than in unprotected animals. In addition, peptide-based epitope mapping of serum antibodies from immunized Aotus showed distinct differences in epitope specificities between protected and unprotected animals.
Collapse
Affiliation(s)
- David R. Cavanagh
- Institute of Immunology and Infection Research, Center for Immunity, Infection and Evolution, Ashworth Laboratories, University of Edinburgh, Edinburgh, United Kingdom
| | - Clemens H. M. Kocken
- Biomedical Primate Research Center, Department of Parasitology, Rijswijk, The Netherlands
| | - John H. White
- Institute of Immunology and Infection Research, Center for Immunity, Infection and Evolution, Ashworth Laboratories, University of Edinburgh, Edinburgh, United Kingdom
| | - Graeme J. M. Cowan
- Institute of Immunology and Infection Research, Center for Immunity, Infection and Evolution, Ashworth Laboratories, University of Edinburgh, Edinburgh, United Kingdom
| | - Kay Samuel
- Scottish National Blood Transfusion Service, Cell Therapy Group, University of Edinburgh, Edinburgh, United Kingdom
| | - Martin A. Dubbeld
- Biomedical Primate Research Center, Department of Parasitology, Rijswijk, The Netherlands
| | | | - Alan W. Thomas
- Biomedical Primate Research Center, Department of Parasitology, Rijswijk, The Netherlands
| | - Jana S. McBride
- Institute of Immunology and Infection Research, Center for Immunity, Infection and Evolution, Ashworth Laboratories, University of Edinburgh, Edinburgh, United Kingdom
| | - David E. Arnot
- Institute of Immunology and Infection Research, Center for Immunity, Infection and Evolution, Ashworth Laboratories, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
49
|
Sequential processing of merozoite surface proteins during and after erythrocyte invasion by Plasmodium falciparum. Infect Immun 2013; 82:924-36. [PMID: 24218484 DOI: 10.1128/iai.00866-13] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Plasmodium falciparum causes malaria disease during the asexual blood stages of infection when merozoites invade erythrocytes and replicate. Merozoite surface proteins (MSPs) are proposed to play a role in the initial binding of merozoites to erythrocytes, but precise roles remain undefined. Based on electron microscopy studies of invading Plasmodium merozoites, it is proposed that the majority of MSPs are cleaved and shed from the surface during invasion, perhaps to release receptor-ligand interactions. In this study, we demonstrate that there is not universal cleavage of MSPs during invasion. Instead, there is sequential and coordinated cleavage and shedding of proteins, indicating a diversity of roles for surface proteins during and after invasion. While MSP1 and peripheral surface proteins such as MSP3, MSP7, serine repeat antigen 4 (SERA4), and SERA5 are cleaved and shed at the tight junction between the invading merozoite and erythrocyte, the glycosylphosphatidylinositol (GPI)-anchored proteins MSP2 and MSP4 are carried into the erythrocyte without detectable processing. Following invasion, MSP2 rapidly degrades within 10 min, whereas MSP4 is maintained for hours. This suggests that while some proteins that are shed upon invasion may have roles in initial contact steps, others function during invasion and are then rapidly degraded, whereas others are internalized for roles during intraerythrocytic development. Interestingly, anti-MSP2 antibodies did not inhibit invasion and instead were carried into erythrocytes and maintained for approximately 20 h without inhibiting parasite development. These findings provide new insights into the mechanisms of invasion and knowledge to advance the development of new drugs and vaccines against malaria.
Collapse
|
50
|
Pusic K, Clements D, Kobuch S, Hui G. Antibody and T cell responses in reciprocal prime-boost studies with full-length and truncated merozoite surface protein 1-42 vaccines. PLoS One 2013; 8:e75939. [PMID: 24098747 PMCID: PMC3786974 DOI: 10.1371/journal.pone.0075939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/16/2013] [Indexed: 11/18/2022] Open
Abstract
The P. falciparum Merozoite Surface Protein 1-42 (MSP1-42) is one of the most studied malaria subunit vaccine candidates. The N-terminal fragment of MSP1-42, MSP1-33, is primarily composed of allelic sequences, and has been shown to possess T helper epitopes that influence protective antibody responses toward the C-terminal region, MSP1-19. A truncated MSP1-42 vaccine, Construct 33-I, consisting of exclusively conserved T epitope regions of MSP1-33 expressed in tandem with MSP1-19, was previously shown to be a more effective immunogen than the full-length MSP1-42 vaccine. Here, by way of reciprocal priming/boosting immunization regimens, we studied the immunogenicity of Construct 33-I in the context of recognition by immune responses induced by the full-length native MSP1-42 protein, in order to gauge the effects of pre- and post-exposures to MSP1-42 on vaccine induced responses. Judging by immune responsiveness, antibody and T cell responses, Construct 33-I was effective as the priming antigen followed by full-length MSP1-42 boosting, as well as the boosting antigen following full-length MSP1-42 priming. In particular, Construct 33-I priming elicited the broadest responsiveness in immunized animals subsequently exposed to MSP1-42. Moreover, Construct 33-I, with its conserved MSP1-33 specific T cell epitopes, was equally well recognized by homologous and heterologous allelic forms of MSP1-42. Serum antibodies raised against Construct 33-I efficiently inhibited the growth of parasites carrying the heterologous MSP1-42 allele. These results suggest that Construct 33-I maintains and/or enhances its immunogenicity in an allelic or strain transcending fashion when deployed in populations having prior or subsequent exposures to native MSP1-42s.
Collapse
Affiliation(s)
- Kae Pusic
- University of Hawaii, School of Medicine, Department of Tropical Medicine, Honolulu, Hawaii, United States of America
- * E-mail:
| | - Danielle Clements
- University of Hawaii, School of Medicine, Department of Tropical Medicine, Honolulu, Hawaii, United States of America
| | - Sophie Kobuch
- University of Hawaii, School of Medicine, Department of Tropical Medicine, Honolulu, Hawaii, United States of America
| | - George Hui
- University of Hawaii, School of Medicine, Department of Tropical Medicine, Honolulu, Hawaii, United States of America
| |
Collapse
|