1
|
Ilani P, Nyarko PB, Camara A, Amenga-Etego LN, Aniweh Y. PfRH5 vaccine; from the bench to the vial. NPJ Vaccines 2025; 10:82. [PMID: 40274841 PMCID: PMC12022022 DOI: 10.1038/s41541-025-01137-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
The search for potent malaria vaccine candidate has seen several twists and turns. Here, we provide a perspective on the current state of PfRH5-based malaria vaccine development, the progress, existing challenges, and future research directions. We discuss the clinical trials in endemic regions, immune correlates of protection, prospects of integrating PfRH5 into multi-antigen vaccine strategies and considerations on the onward development/deployment of PfRH5 vaccine from the laboratory to endemic communities.
Collapse
Affiliation(s)
- Philip Ilani
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Prince B Nyarko
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
- Laboratory of Pathogens and Host Immunity (LPHI), CNRS, University of Montpellier, Montpellier, France
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain
| | - Abdouramane Camara
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Lucas N Amenga-Etego
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana
| | - Yaw Aniweh
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Legon, Accra, Ghana.
| |
Collapse
|
2
|
Devi S, Negi S, Tandel N, Dalai SK, Tyagi RK. Oleuropein: a viable therapeutic option for malaria and cancer. Drug Discov Today 2025; 30:104254. [PMID: 39608487 DOI: 10.1016/j.drudis.2024.104254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024]
Abstract
Oleuropein (OLP) holds promise as a therapeutic candidate for both Plasmodium falciparum infection and cancer. It modulates the phosphoinositide 3-kinase (PI3K)-Akt1 signaling pathway to regulate inflammation and restore immune homeostasis. Moreover, it influences the cell death/autophagy axis, along with increasing the antimalarial efficacy of artemisinin. Our findings indicate that the anti-breast-cancer effect of OLP could be mediated by regulating the balance of T helper 17 and regulatory T cells. Additionally, we discuss the use of hematopoietic-stem-cell-transplanted immunodeficient mice with a humanized immune system for validating the antimalarial activity, autophagy and anticancer activity of OLP.
Collapse
Affiliation(s)
- Sonia Devi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India
| | - Sushmita Negi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India
| | - Nikunj Tandel
- CSIR-Centre For Cellular & Molecular Biology (CCMB), Hyderabad, Telangana 500007, India
| | - Sarat K Dalai
- Institute of Science, Nirma University, SG Highway, Gujarat 382481, India
| | - Rajeev K Tyagi
- Division of Cell Biology and Immunology, Biomedical Parasitology and Translational-immunology Lab, CSIR-Institute of Microbial Technology (IMTECH), Chandigarh 160036, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
3
|
Patel AK, Trageser K, Kim H, Lim W, Adler C, Porter B, Ni M, Wei Y, Atwal GS, Bigdelou P, Kulshreshtha V, Ajithdoss D, Zhong J, Tu N, Macdonald L, Murphy A, Frleta D. Peripheral human red blood cell development in human immune system mouse model with heme oxygenase-1 deficiency. Blood Adv 2024; 8:5975-5987. [PMID: 39348688 PMCID: PMC11629214 DOI: 10.1182/bloodadvances.2023011754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 07/17/2024] [Accepted: 09/10/2024] [Indexed: 10/02/2024] Open
Abstract
ABSTRACT A challenge for human immune system (HIS) mouse models has been the lack of human red blood cell (hRBC) survival after engraftment of these immune-deficient mice with human CD34+ hematopoietic stem cells (HSCs). This limits the use of HIS models for preclinical testing of targets directed at hRBC-related diseases. Although human white blood cells can develop in the peripheral blood of mice engrafted with human HSCs, peripheral hRBCs are quickly phagocytosed by murine macrophages upon egress from the bone marrow. Genetic ablation of murine myeloid cells results in severe pathology in resulting mice, rendering such an approach to increase hRBC survival in HIS mice impractical. Heme oxygenase-1 (HMOX-1)-deficient mice have reduced macrophages due to toxic buildup of intracellular heme upon engulfment of RBCs, but do not have an overall loss of myeloid cells. We took advantage of this observation and generated HMOX-1-/- mice on a humanized M-CSF/SIRPα/CD47 Rag2-/- IL-2Rγ-/- background. These mice have reduced murine macrophages but comparable levels of murine myeloid cells to HMOX-1+/+ control mice in the same background. Injected hRBCs survive longer in HMOX-1-/- mice than in HMOX-1+/+ controls. Additionally, upon human HSC engraftment, hRBCs can be observed in the peripheral blood of HMOX-1-/- humanized M-CSF/SIRPα/CD47 Rag2-/- IL-2Rγ-/- mice, and hRBC levels can be increased by treatment with human erythropoietin. Given that hRBC are present in the peripheral blood of engrafted HMOX-1-/- mice, these mice have the potential to be used for hematologic disease modeling, and for testing therapeutic treatments for hRBC diseases in vivo.
Collapse
Affiliation(s)
| | | | - Hyunjin Kim
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY
| | - Weikeat Lim
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY
| | | | | | - Min Ni
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY
| | - Yi Wei
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY
| | | | | | | | | | - Jun Zhong
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY
| | - Naxin Tu
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY
| | | | | | | |
Collapse
|
4
|
Miyazaki Y, Miyazaki S. Reporter parasite lines: valuable tools for the study of Plasmodium biology. Trends Parasitol 2024; 40:1000-1015. [PMID: 39389901 DOI: 10.1016/j.pt.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024]
Abstract
The human malaria parasite Plasmodium falciparum causes the most severe form of malaria in endemic regions and is transmitted via mosquito bites. To better understand the biology of this deadly pathogen, a variety of P. falciparum reporter lines have been generated using transgenic approaches to express reporter proteins, such as fluorescent proteins and luciferases. This review discusses the advances in recently generated P. falciparum transgenic reporter lines, which will aid in the investigation of parasite physiology and the discovery of novel antimalarial drugs. Future prospects for the generation of new and superior human malaria parasite reporter lines are also discussed, and unresolved questions in malaria biology are highlighted to help boost support for the development and implementation of malaria treatments.
Collapse
Affiliation(s)
- Yukiko Miyazaki
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Shinya Miyazaki
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.
| |
Collapse
|
5
|
Barrett JR, Pipini D, Wright ND, Cooper AJR, Gorini G, Quinkert D, Lias AM, Davies H, Rigby CA, Aleshnick M, Williams BG, Bradshaw WJ, Paterson NG, Martinson T, Kirtley P, Picard L, Wiggins CD, Donnellan FR, King LDW, Wang LT, Popplewell JF, Silk SE, de Ruiter Swain J, Skinner K, Kotraiah V, Noe AR, MacGill RS, King CR, Birkett AJ, Soisson LA, Minassian AM, Lauffenburger DA, Miura K, Long CA, Wilder BK, Koekemoer L, Tan J, Nielsen CM, McHugh K, Draper SJ. Analysis of the diverse antigenic landscape of the malaria protein RH5 identifies a potent vaccine-induced human public antibody clonotype. Cell 2024; 187:4964-4980.e21. [PMID: 39059380 PMCID: PMC11380582 DOI: 10.1016/j.cell.2024.06.015] [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: 09/28/2023] [Revised: 04/14/2024] [Accepted: 06/10/2024] [Indexed: 07/28/2024]
Abstract
The highly conserved and essential Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) has emerged as the leading target for vaccines against the disease-causing blood stage of malaria. However, the features of the human vaccine-induced antibody response that confer highly potent inhibition of malaria parasite invasion into red blood cells are not well defined. Here, we characterize 236 human IgG monoclonal antibodies, derived from 15 donors, induced by the most advanced PfRH5 vaccine. We define the antigenic landscape of this molecule and establish that epitope specificity, antibody association rate, and intra-PfRH5 antibody interactions are key determinants of functional anti-parasitic potency. In addition, we identify a germline IgG gene combination that results in an exceptionally potent class of antibody and demonstrate its prophylactic potential to protect against P. falciparum parasite challenge in vivo. This comprehensive dataset provides a framework to guide rational design of next-generation vaccines and prophylactic antibodies to protect against blood-stage malaria.
Collapse
Affiliation(s)
- Jordan R Barrett
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Dimitra Pipini
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Nathan D Wright
- Centre for Medicines Discovery, University of Oxford, Oxford OX3 7FZ, UK
| | - Andrew J R Cooper
- Antibody Biology Unit, Laboratory of Immunogenetics, NIAID/NIH, Rockville, MD 20852, USA
| | - Giacomo Gorini
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Doris Quinkert
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Amelia M Lias
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Hannah Davies
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Cassandra A Rigby
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Maya Aleshnick
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Barnabas G Williams
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - William J Bradshaw
- Centre for Medicines Discovery, University of Oxford, Oxford OX3 7FZ, UK
| | - Neil G Paterson
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, Oxfordshire OX11 0DE, UK
| | - Thomas Martinson
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Payton Kirtley
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Luc Picard
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | | | - Francesca R Donnellan
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Lloyd D W King
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Lawrence T Wang
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; Antibody Biology Unit, Laboratory of Immunogenetics, NIAID/NIH, Rockville, MD 20852, USA
| | | | - Sarah E Silk
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Jed de Ruiter Swain
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Katherine Skinner
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Amy R Noe
- Leidos Life Sciences, Frederick, MD, USA
| | - Randall S MacGill
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA
| | - C Richter King
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA
| | - Ashley J Birkett
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA
| | | | - Angela M Minassian
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | | | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Brandon K Wilder
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Lizbé Koekemoer
- Centre for Medicines Discovery, University of Oxford, Oxford OX3 7FZ, UK
| | - Joshua Tan
- Antibody Biology Unit, Laboratory of Immunogenetics, NIAID/NIH, Rockville, MD 20852, USA
| | - Carolyn M Nielsen
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Kirsty McHugh
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK; The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
6
|
Wang LT, Cooper AJR, Farrell B, Miura K, Diouf A, Müller-Sienerth N, Crosnier C, Purser L, Kirtley PJ, Maciuszek M, Barrett JR, McHugh K, Ogwang R, Tucker C, Li S, Doumbo S, Doumtabe D, Pyo CW, Skinner J, Nielsen CM, Silk SE, Kayentao K, Ongoiba A, Zhao M, Nguyen DC, Lee FEH, Minassian AM, Geraghty DE, Traore B, Seder RA, Wilder BK, Crompton PD, Wright GJ, Long CA, Draper SJ, Higgins MK, Tan J. Natural malaria infection elicits rare but potent neutralizing antibodies to the blood-stage antigen RH5. Cell 2024; 187:4981-4995.e14. [PMID: 39059381 PMCID: PMC11383431 DOI: 10.1016/j.cell.2024.06.037] [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: 09/15/2023] [Revised: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024]
Abstract
Plasmodium falciparum reticulocyte-binding protein homolog 5 (RH5) is the most advanced blood-stage malaria vaccine candidate and is being evaluated for efficacy in endemic regions, emphasizing the need to study the underlying antibody response to RH5 during natural infection, which could augment or counteract responses to vaccination. Here, we found that RH5-reactive B cells were rare, and circulating immunoglobulin G (IgG) responses to RH5 were short-lived in malaria-exposed Malian individuals, despite repeated infections over multiple years. RH5-specific monoclonal antibodies isolated from eight malaria-exposed individuals mostly targeted non-neutralizing epitopes, in contrast to antibodies isolated from five RH5-vaccinated, malaria-naive UK individuals. However, MAD8-151 and MAD8-502, isolated from two malaria-exposed Malian individuals, were among the most potent neutralizers out of 186 antibodies from both cohorts and targeted the same epitopes as the most potent vaccine-induced antibodies. These results suggest that natural malaria infection may boost RH5-vaccine-induced responses and provide a clear strategy for the development of next-generation RH5 vaccines.
Collapse
Affiliation(s)
- Lawrence T Wang
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Medical Scientist Training Program, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Andrew J R Cooper
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Brendan Farrell
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | | | - Cécile Crosnier
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK
| | - Lauren Purser
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Payton J Kirtley
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97006, USA
| | - Maciej Maciuszek
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Jordan R Barrett
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Kirsty McHugh
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Rodney Ogwang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Courtney Tucker
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Shanping Li
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Safiatou Doumbo
- Mali International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Point G, BP 1805 Bamako, Mali
| | - Didier Doumtabe
- Mali International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Point G, BP 1805 Bamako, Mali
| | - Chul-Woo Pyo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jeff Skinner
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Carolyn M Nielsen
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Sarah E Silk
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Kassoum Kayentao
- Mali International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Point G, BP 1805 Bamako, Mali
| | - Aissata Ongoiba
- Mali International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Point G, BP 1805 Bamako, Mali
| | - Ming Zhao
- Protein Chemistry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Doan C Nguyen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA 30322, USA
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA 30322, USA
| | - Angela M Minassian
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| | - Daniel E Geraghty
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Boubacar Traore
- Mali International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, Point G, BP 1805 Bamako, Mali
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brandon K Wilder
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97006, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Gavin J Wright
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Joshua Tan
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|
7
|
Robarts DR, Paine-Cabrera D, Kotulkar M, Venneman KK, Gunewardena S, Foquet L, Bial G, Apte U. Identifying novel mechanisms of per- and polyfluoroalkyl substance-induced hepatotoxicity using FRG humanized mice. Arch Toxicol 2024; 98:3063-3075. [PMID: 38782768 DOI: 10.1007/s00204-024-03789-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) such as perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) and perfluoro-2-methyl-3-oxahexanoic acid (GenX), the new replacement PFAS, are major environmental contaminants. In rodents, these PFAS induce several adverse effects on the liver, including increased proliferation, hepatomegaly, steatosis, hypercholesterolemia, nonalcoholic fatty liver disease and liver cancers. Activation of peroxisome proliferator receptor alpha by PFAS is considered the primary mechanism of action in rodent hepatocyte-induced proliferation. However, the human relevance of this mechanism is uncertain. We investigated human-relevant mechanisms of PFAS-induced adverse hepatic effects using FRG liver-chimeric humanized mice with livers repopulated with functional human hepatocytes. Male FRG humanized mice were treated with 0.067 mg/L of PFOA, 0.145 mg/L of PFOS, or 1 mg/L of GenX in drinking water for 28 days. PFOS caused a significant decrease in total serum cholesterol and LDL/VLDL, whereas GenX caused a significant elevation in LDL/VLDL with no change in total cholesterol and HDL. All three PFAS induced significant hepatocyte proliferation. RNA-sequencing with alignment to the human genome showed a total of 240, 162, and 619 differentially expressed genes after PFOA, PFOS, and GenX exposure, respectively. Upstream regulator analysis revealed that all three PFAS induced activation of p53 and inhibition of androgen receptor and NR1D1, a transcriptional repressor important in circadian rhythm. Further biochemical studies confirmed NR1D1 inhibition and in silico modeling indicated potential interaction of all three PFAS with the DNA-binding domain of NR1D1. In conclusion, our studies using FRG humanized mice have revealed new human-relevant molecular mechanisms of PFAS including their previously unknown effect on circadian rhythm.
Collapse
Affiliation(s)
- Dakota R Robarts
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MS1018, Kansas City, KS, 66160, USA
| | - Diego Paine-Cabrera
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MS1018, Kansas City, KS, 66160, USA
| | - Manasi Kotulkar
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MS1018, Kansas City, KS, 66160, USA
| | - Kaitlyn K Venneman
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MS1018, Kansas City, KS, 66160, USA
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Greg Bial
- Yecuris Corporation, Tualatin, OR, USA
| | - Udayan Apte
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MS1018, Kansas City, KS, 66160, USA.
| |
Collapse
|
8
|
King LDW, Pulido D, Barrett JR, Davies H, Quinkert D, Lias AM, Silk SE, Pattinson DJ, Diouf A, Williams BG, McHugh K, Rodrigues A, Rigby CA, Strazza V, Suurbaar J, Rees-Spear C, Dabbs RA, Ishizuka AS, Zhou Y, Gupta G, Jin J, Li Y, Carnrot C, Minassian AM, Campeotto I, Fleishman SJ, Noe AR, MacGill RS, King CR, Birkett AJ, Soisson LA, Long CA, Miura K, Ashfield R, Skinner K, Howarth MR, Biswas S, Draper SJ. Preclinical development of a stabilized RH5 virus-like particle vaccine that induces improved antimalarial antibodies. Cell Rep Med 2024; 5:101654. [PMID: 39019011 PMCID: PMC11293324 DOI: 10.1016/j.xcrm.2024.101654] [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: 01/05/2024] [Revised: 04/12/2024] [Accepted: 06/19/2024] [Indexed: 07/19/2024]
Abstract
Plasmodium falciparum reticulocyte-binding protein homolog 5 (RH5) is a leading blood-stage malaria vaccine antigen target, currently in a phase 2b clinical trial as a full-length soluble protein/adjuvant vaccine candidate called RH5.1/Matrix-M. We identify that disordered regions of the full-length RH5 molecule induce non-growth inhibitory antibodies in human vaccinees and that a re-engineered and stabilized immunogen (including just the alpha-helical core of RH5) induces a qualitatively superior growth inhibitory antibody response in rats vaccinated with this protein formulated in Matrix-M adjuvant. In parallel, bioconjugation of this immunogen, termed "RH5.2," to hepatitis B surface antigen virus-like particles (VLPs) using the "plug-and-display" SpyTag-SpyCatcher platform technology also enables superior quantitative antibody immunogenicity over soluble protein/adjuvant in vaccinated mice and rats. These studies identify a blood-stage malaria vaccine candidate that may improve upon the current leading soluble protein vaccine candidate RH5.1/Matrix-M. The RH5.2-VLP/Matrix-M vaccine candidate is now under evaluation in phase 1a/b clinical trials.
Collapse
Affiliation(s)
- Lloyd D W King
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - David Pulido
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Jordan R Barrett
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Hannah Davies
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Doris Quinkert
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Amelia M Lias
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Sarah E Silk
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - David J Pattinson
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Barnabas G Williams
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Kirsty McHugh
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Ana Rodrigues
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK
| | - Cassandra A Rigby
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK
| | - Veronica Strazza
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK
| | - Jonathan Suurbaar
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK; West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Accra LG 54, Ghana
| | - Chloe Rees-Spear
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK; London School of Hygiene and Tropical Medicine, WC1E 7HT London, UK
| | - Rebecca A Dabbs
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Andrew S Ishizuka
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Yu Zhou
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Gaurav Gupta
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Jing Jin
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Yuanyuan Li
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | | | - Angela M Minassian
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Ivan Campeotto
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK
| | - Sarel J Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Amy R Noe
- Leidos Life Sciences, Frederick, MD, USA
| | - Randall S MacGill
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA
| | - C Richter King
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA
| | - Ashley J Birkett
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA
| | | | - 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
| | - Rebecca Ashfield
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Katherine Skinner
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Mark R Howarth
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK
| | - Sumi Biswas
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, OX1 3QU Oxford, UK; Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, OX1 3QU Oxford, UK; The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
9
|
Goswami D, Patel H, Betz W, Armstrong J, Camargo N, Patil A, Chakravarty S, Murphy SC, Sim BKL, Vaughan AM, Hoffman SL, Kappe SH. A replication competent Plasmodium falciparum parasite completely attenuated by dual gene deletion. EMBO Mol Med 2024; 16:723-754. [PMID: 38514791 PMCID: PMC11018819 DOI: 10.1038/s44321-024-00057-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Vaccination with infectious Plasmodium falciparum (Pf) sporozoites (SPZ) administered with antimalarial drugs (PfSPZ-CVac), confers superior sterilizing protection against infection when compared to vaccination with replication-deficient, radiation-attenuated PfSPZ. However, the requirement for drug administration constitutes a major limitation for PfSPZ-CVac. To obviate this limitation, we generated late liver stage-arresting replication competent (LARC) parasites by deletion of the Mei2 and LINUP genes (mei2-/linup- or LARC2). We show that Plasmodium yoelii (Py) LARC2 sporozoites did not cause breakthrough blood stage infections and engendered durable sterilizing immunity against various infectious sporozoite challenges in diverse strains of mice. We next genetically engineered a PfLARC2 parasite strain that was devoid of extraneous DNA and produced cryopreserved PfSPZ-LARC2. PfSPZ-LARC2 liver stages replicated robustly in liver-humanized mice but displayed severe defects in late liver stage differentiation and did not form liver stage merozoites. This resulted in complete abrogation of parasite transition to viable blood stage infection. Therefore, PfSPZ-LARC2 is the next-generation vaccine strain expected to unite the safety profile of radiation-attenuated PfSPZ with the superior protective efficacy of PfSPZ-CVac.
Collapse
Affiliation(s)
- Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Hardik Patel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - William Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Janna Armstrong
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
| | - Asha Patil
- Sanaria Inc., 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | | | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - B Kim Lee Sim
- Sanaria Inc., 9800 Medical Center Dr., Rockville, MD, 20850, USA
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Stefan Hi Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA, 98109, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
10
|
Ma M, Ge JY, Nie YZ, Li YM, Zheng YW. Developing Humanized Animal Models with Transplantable Human iPSC-Derived Cells. FRONT BIOSCI-LANDMRK 2024; 29:34. [PMID: 38287837 DOI: 10.31083/j.fbl2901034] [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: 09/16/2023] [Revised: 12/02/2023] [Accepted: 12/22/2023] [Indexed: 01/31/2024]
Abstract
Establishing reliable and reproducible animal models for disease modelling, drug screening and the understanding of disease susceptibility and pathogenesis is critical. However, traditional animal models differ significantly from humans in terms of physiology, immune response, and pathogenesis. As a result, it is difficult to translate laboratory findings into biomedical applications. Although several animal models with human chimeric genes, organs or systems have been developed in the past, their limited engraftment rate and physiological functions are a major obstacle to realize convincing models of humans. The lack of human transplantation resources and insufficient immune tolerance of recipient animals are the main challenges that need to be overcome to generate fully humanized animals. Recent advances in gene editing and pluripotent stem cell-based xenotransplantation technologies offer opportunities to create more accessible human-like models for biomedical research. In this article, we have combined our laboratory expertise to summarize humanized animal models, with a focus on hematopoietic/immune system and liver. We discuss their generation strategies and the potential donor cell sources, with particular attention given to human pluripotent stem cells. In particular, we discuss the advantages, limitations and emerging trends in their clinical and pharmaceutical applications. By providing insights into the current state of humanized animal models and their potential for biomedical applications, this article aims to advance the development of more accurate and reliable animal models for disease modeling and drug screening.
Collapse
Affiliation(s)
- Min Ma
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212001 Zhenjiang, Jiangsu, China
| | - Jian-Yun Ge
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, 529020 Jiangmen, Guangdong, China
| | - Yun-Zhong Nie
- Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 108-8639 Tokyo, Japan
| | - Yu-Mei Li
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212001 Zhenjiang, Jiangsu, China
| | - Yun-Wen Zheng
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, 212001 Zhenjiang, Jiangsu, China
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, 529020 Jiangmen, Guangdong, China
- Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, 108-8639 Tokyo, Japan
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 278-8510 Noda, Japan
| |
Collapse
|
11
|
Browne DJ, Kelly AM, Brady J, Proietti C, Sarathkumara YD, Pattinson DJ, Doolan DL. Evaluating the stability of host-reference gene expression and simultaneously quantifying parasite burden and host immune responses in murine malaria. Sci Rep 2023; 13:21071. [PMID: 38030676 PMCID: PMC10687243 DOI: 10.1038/s41598-023-48066-9] [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: 09/06/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023] Open
Abstract
The efficacy of pre-erythrocytic stage malaria antigens or vaccine platforms is routinely assessed in murine models challenged with Plasmodium sporozoites. Relative liver-stage parasite burden is quantified using reverse transcription quantitative PCR (RTqPCR), which relies on constitutively expressed endogenous control reference genes. However, the stability of host-reference gene expression for RTqPCR analysis following Plasmodium challenge and immunization has not been systematically evaluated. Herein, we evaluated the stability of expression of twelve common RTqPCR reference genes in a murine model of Plasmodium yoelii sporozoite challenge and DNA-adenovirus IV 'Prime-Target' immunization. Significant changes in expression for six of twelve reference genes were shown by one-way ANOVA, when comparing gene expression levels among challenge, immunized, and naïve mice groups. These changes were attributed to parasite challenge or immunization when comparing group means using post-hoc Bonferroni corrected multiple comparison testing. Succinate dehydrogenase (SDHA) and TATA-binding protein (TBP) were identified as stable host-reference genes suitable for relative RTqPCR data normalisation, using the RefFinder package. We defined a robust threshold of 'partial-protection' with these genes and developed a strategy to simultaneously quantify matched host parasite burden and cytokine responses following immunisation or challenge. This is the first report systematically identifying reliable host reference genes for RTqPCR analysis following Plasmodium sporozoite challenge. A robust RTqPCR protocol incorporating reliable reference genes which enables simultaneous analysis of host whole-liver cytokine responses and parasite burden will significantly standardise and enhance results between international malaria vaccine efficacy studies.
Collapse
Affiliation(s)
- Daniel J Browne
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4870, Australia
| | - Ashton M Kelly
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4870, Australia
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd, St Lucia, QLD, 4072, Australia
| | - Jamie Brady
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4870, Australia
| | - Carla Proietti
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd, St Lucia, QLD, 4072, Australia
| | - Yomani D Sarathkumara
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4870, Australia
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd, St Lucia, QLD, 4072, Australia
| | - David J Pattinson
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4870, Australia
| | - Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4870, Australia.
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd, St Lucia, QLD, 4072, Australia.
- Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4870, Australia.
| |
Collapse
|
12
|
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: 6.0] [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
|
13
|
van Niekerk DD, du Toit F, Green K, Palm D, Snoep JL. A detailed kinetic model of glycolysis in Plasmodium falciparum-infected red blood cells for antimalarial drug target identification. J Biol Chem 2023; 299:105111. [PMID: 37517694 PMCID: PMC10474083 DOI: 10.1016/j.jbc.2023.105111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/11/2023] [Accepted: 07/23/2023] [Indexed: 08/01/2023] Open
Abstract
Upon infection by the malaria parasite Plasmodium falciparum, the glycolytic rate of a red blood cell increases up to 100-fold, possibly contributing to lactic acidosis and hypoglycemia in patients with severe malaria. This dramatic increase in glucose uptake and metabolism was correctly predicted by a newly constructed detailed enzyme kinetic model of glucose metabolism in the trophozoite-infected red blood cell. Subsequently, we expanded the model to simulate an infected red blood cell culture, including the different asexual blood-stage forms of the malaria parasite. The model simulations were in good agreement with experimental data, for which the measured parasitic volume was an important parameter. Upon further analysis of the model, we identified glucose transport as a drug target that would specifically affect infected red blood cells, which was confirmed experimentally with inhibitor titrations. This model can be a first step in constructing a whole-body model for glucose metabolism in malaria patients to evaluate the contribution of the parasite's metabolism to the disease state.
Collapse
Affiliation(s)
- David D van Niekerk
- Department of Biochemistry, Stellenbosch University, Matieland, South Africa
| | - Francois du Toit
- Department of Biochemistry, Stellenbosch University, Matieland, South Africa
| | - Kathleen Green
- Department of Biochemistry, Stellenbosch University, Matieland, South Africa
| | - Danie Palm
- Department of Biochemistry, Stellenbosch University, Matieland, South Africa
| | - Jacky L Snoep
- Department of Biochemistry, Stellenbosch University, Matieland, South Africa; Molecular Cell Biology, Vrije Universiteit, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Jiménez-Díaz MB, Möhrle JJ, Angulo-Barturen I, Demarta-Gatsi C. Using Cryopreserved Plasmodium falciparum Sporozoites in a Humanized Mouse Model to Study Early Malaria Infection Processes and Test Prophylactic Treatments. Microorganisms 2023; 11:2209. [PMID: 37764054 PMCID: PMC10536749 DOI: 10.3390/microorganisms11092209] [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: 07/11/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 09/29/2023] Open
Abstract
In addition to vector control, long-lasting insecticidal nets and case management, the prevention of infection through vaccination and/or chemoprevention are playing an increasing role in the drive to eradicate malaria. These preventative approaches represent opportunities for improvement: new drugs may be discovered that target the early infectious stages of the Plasmodium parasite in the liver (rather than the symptomatic, abundant blood stage), and new, exciting vaccination technologies have recently been validated (using mRNA or novel adjuvants). Exploiting these possibilities requires the availability of humanized mouse models that support P. falciparum infection yet avoid the hazardous use of infectious mosquitoes. Here, we show that commercially available P. falciparum sporozoites and FRG mice carrying human hepatocytes and red blood cells faithfully recapitulate the early human malaria disease process, presenting an opportunity to use this model for the evaluation of prophylactic treatments with a novel mode of action.
Collapse
|
15
|
Gehlot P, Vyas VK. Recent advances on patents of Plasmodium falciparum dihydroorotate dehydrogenase ( PfDHODH) inhibitors as antimalarial agents. Expert Opin Ther Pat 2023; 33:579-596. [PMID: 37942637 DOI: 10.1080/13543776.2023.2280596] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
INTRODUCTION Pyrimidine nucleotides are essential for the parasite's growth and replication. Parasites have only a de novo pathway for the biosynthesis of pyrimidine nucleotides. Dihydroorotate dehydrogenase (DHODH) enzyme is involved in the rate-limiting step of the pyrimidine biosynthesis pathway. DHODH is a biochemical target for the discovery of new antimalarial agents. AREA COVERED This review discussed the development of patented PfDHODH inhibitors published between 2007 and 2023 along with their chemical structures and activities. EXPERT OPINION PfDHODH enzyme is involved in the rate-limiting fourth step of the pyrimidine biosynthesis pathway. Thus, inhibition of PfDHODH using species-selective inhibitors has drawn much attention for treating malaria because they inhibit parasite growth without affecting normal human functions. Looking at the current scenario of antimalarial drug resistance with most of the available antimalarial drugs, there is a huge need for targeted newer agents. Newer agents with unique mechanisms of action may be devoid of drug toxicity, adverse effects, and the ability of parasites to quickly gain resistance, and PfDHODH inhibitors can be those newer agents. Many PfDHODH inhibitors were patented in the past, and the dependency of Plasmodium on de novo pyrimidine provided a new approach for the development of novel antimalarial agents.
Collapse
Affiliation(s)
- Pinky Gehlot
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, India
| |
Collapse
|
16
|
Karnik I, Her Z, Neo SH, Liu WN, Chen Q. Emerging Preclinical Applications of Humanized Mouse Models in the Discovery and Validation of Novel Immunotherapeutics and Their Mechanisms of Action for Improved Cancer Treatment. Pharmaceutics 2023; 15:1600. [PMID: 37376049 DOI: 10.3390/pharmaceutics15061600] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer therapeutics have undergone immense research over the past decade. While chemotherapies remain the mainstay treatments for many cancers, the advent of new molecular techniques has opened doors for more targeted modalities towards cancer cells. Although immune checkpoint inhibitors (ICIs) have demonstrated therapeutic efficacy in treating cancer, adverse side effects related to excessive inflammation are often reported. There is a lack of clinically relevant animal models to probe the human immune response towards ICI-based interventions. Humanized mouse models have emerged as valuable tools for pre-clinical research to evaluate the efficacy and safety of immunotherapy. This review focuses on the establishment of humanized mouse models, highlighting the challenges and recent advances in these models for targeted drug discovery and the validation of therapeutic strategies in cancer treatment. Furthermore, the potential of these models in the process of uncovering novel disease mechanisms is discussed.
Collapse
Affiliation(s)
- Isha Karnik
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Zhisheng Her
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Shu Hui Neo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Wai Nam Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| |
Collapse
|
17
|
Robarts DR, Paine-Cabrera D, Kotulkar M, Venneman KK, Gunewardena S, Corton JC, Lau C, Foquet L, Bial G, Apte U. Identifying Human Specific Adverse Outcome Pathways of Per- and Polyfluoroalkyl Substances Using Liver-Chimeric Humanized Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526711. [PMID: 36778348 PMCID: PMC9915685 DOI: 10.1101/2023.02.01.526711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Per- and polyfluoroalkyl substances (PFAS) are persistent organic pollutants with myriad adverse effects. While perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) are the most common contaminants, levels of replacement PFAS, such as perfluoro-2-methyl-3-oxahexanoic acid (GenX), are increasing. In rodents, PFOA, PFOS, and GenX have several adverse effects on the liver, including nonalcoholic fatty liver disease. Objective We aimed to determine human-relevant mechanisms of PFAS induced adverse hepatic effects using FRG liver-chimeric humanized mice with livers repopulated with functional human hepatocytes. Methods Male humanized mice were treated with 0.067 mg/L of PFOA, 0.145 mg/L of PFOS, or 1 mg/L of GenX in drinking water for 28 days. Liver and serum were collected for pathology and clinical chemistry, respectively. RNA-sequencing coupled with pathway analysis was used to determine molecular mechanisms. Results PFOS caused a significant decrease in total serum cholesterol and LDL/VLDL, whereas GenX caused a significant elevation in LDL/VLDL with no change in total cholesterol and HDL. PFOA had no significant changes in serum LDL/VLDL and total cholesterol. All three PFAS induced significant hepatocyte proliferation. RNA-sequencing with alignment to the human genome showed a total of 240, 162, and 619 differentially expressed genes after PFOA, PFOS, and GenX exposure, respectively. Upstream regulator analysis revealed inhibition of NR1D1, a transcriptional repressor important in circadian rhythm, as the major common molecular change in all PFAS treatments. PFAS treated mice had significant nuclear localization of NR1D1. In silico modeling showed PFOA, PFOS, and GenX potentially interact with the DNA-binding domain of NR1D1. Discussion These data implicate PFAS in circadian rhythm disruption via inhibition of NR1D1. These studies show that FRG humanized mice are a useful tool for studying the adverse outcome pathways of environmental pollutants on human hepatocytes in situ.
Collapse
Affiliation(s)
- Dakota R. Robarts
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Diego Paine-Cabrera
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Manasi Kotulkar
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Kaitlyn K. Venneman
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| | - J. Christopher Corton
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. EPA, Research Triangle Park, NC
| | - Christopher Lau
- Center for Public Health and Environmental Assessment, Office of Research and Development, US EPA, Research Triangle Park, NC
| | | | | | - Udayan Apte
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
18
|
Cherkaoui‐Rbati MH, Andenmatten N, Burgert L, Egbelowo OF, Fendel R, Fornari C, Gabel M, Ward J, Möhrle JJ, Gobeau N. A pharmacokinetic-pharmacodynamic model for chemoprotective agents against malaria. CPT Pharmacometrics Syst Pharmacol 2022; 12:50-61. [PMID: 36412499 PMCID: PMC9835136 DOI: 10.1002/psp4.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 08/28/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
Chemoprophylactics are a vital tool in the fight against malaria. They can be used to protect populations at risk, such as children younger than the age of 5 in areas of seasonal malaria transmission or pregnant women. Currently approved chemoprophylactics all present challenges. There are either concerns about unacceptable adverse effects such as neuropsychiatric sequalae (mefloquine), risks of hemolysis in patients with G6PD deficiency (8-aminoquinolines such as tafenoquine), or cost and daily dosing (atovaquone-proguanil). Therefore, there is a need to develop new chemoprophylactic agents to provide more affordable therapies with better compliance through improving properties such as pharmacokinetics to allow weekly, preferably monthly, dosing. Here we present a pharmacokinetic-pharmacodynamic (PKPD) model constructed using DSM265 (a dihydroorotate dehydrogenase inhibitor with activity against the liver schizonts of malaria, therefore, a prophylaxis candidate). The PKPD model mimics the parasite lifecycle by describing parasite dynamics and drug activity during the liver and blood stages. A major challenge is the estimation of model parameters, as only blood-stage parasites can be observed once they have reached a threshold. By combining qualitative and quantitative knowledge about the parasite from various sources, it has been shown that it is possible to infer information about liver-stage growth and its initial infection level. Furthermore, by integrating clinical data, the killing effect of the drug on liver- and blood-stage parasites can be included in the PKPD model, and a clinical outcome can be predicted. Despite multiple challenges, the presented model has the potential to help translation from preclinical to late development for new chemoprophylactic candidates.
Collapse
Affiliation(s)
- Mohammed H. Cherkaoui‐Rbati
- Medicines for Malaria VentureGenevaSwitzerland,Present address:
Pharmaceutical Sciences, Pharma Research and Early DevelopmentHoffmann‐La RocheBaselSwitzerland
| | - Nicole Andenmatten
- Medicines for Malaria VentureGenevaSwitzerland,Present address:
Lonza AGVispSwitzerland
| | | | - Oluwaseun F. Egbelowo
- Division of Clinical Pharmacology, Department of MedicineThe University of Texas at AustinTexasAustinUSA
| | - Rolf Fendel
- Institute for Tropical MedicineUniversity of TübingenTübingenGermany
| | | | - Michael Gabel
- Center for Modelling and Simulation in the Biosciences, BioQuant‐CenterUniversity of HeidelbergHeidelbergGermany
| | - John Ward
- Department of Mathematical SciencesUniversity of LoughboroughLoughboroughUK
| | | | | |
Collapse
|
19
|
Valenciano AL, Gomez-Lorenzo MG, Vega-Rodríguez J, Adams JH, Roth A. In vitro models for human malaria: targeting the liver stage. Trends Parasitol 2022; 38:758-774. [PMID: 35780012 PMCID: PMC9378454 DOI: 10.1016/j.pt.2022.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
The Plasmodium liver stage represents a vulnerable therapeutic target to prevent disease progression as the parasite resides in the liver before clinical representation caused by intraerythrocytic development. However, most antimalarial drugs target the blood stage of the parasite's life cycle, and the few drugs that target the liver stage are lethal to patients with a glucose-6-phosphate dehydrogenase deficiency. Furthermore, implementation of in vitro liver models to study and develop novel therapeutics against the liver stage of human Plasmodium species remains challenging. In this review, we focus on the progression of in vitro liver models developed for human Plasmodium spp. parasites, provide a brief review on important assay requirements, and lastly present recommendations to improve models to enhance the discovery process of novel preclinical therapeutics.
Collapse
Affiliation(s)
- Ana Lisa Valenciano
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA; Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Maria G Gomez-Lorenzo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
20
|
Aparici Herraiz I, Caires HR, Castillo-Fernández Ó, Sima N, Méndez-Mora L, Risueño RM, Sattabongkot J, Roobsoong W, Hernández-Machado A, Fernandez-Becerra C, Barrias CC, del Portillo HA. Advancing Key Gaps in the Knowledge of Plasmodium vivax Cryptic Infections Using Humanized Mouse Models and Organs-on-Chips. Front Cell Infect Microbiol 2022; 12:920204. [PMID: 35873153 PMCID: PMC9302440 DOI: 10.3389/fcimb.2022.920204] [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: 04/14/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium vivax is the most widely distributed human malaria parasite representing 36.3% of disease burden in the South-East Asia region and the most predominant species in the region of the Americas. Recent estimates indicate that 3.3 billion of people are under risk of infection with circa 7 million clinical cases reported each year. This burden is certainly underestimated as the vast majority of chronic infections are asymptomatic. For centuries, it has been widely accepted that the only source of cryptic parasites is the liver dormant stages known as hypnozoites. However, recent evidence indicates that niches outside the liver, in particular in the spleen and the bone marrow, can represent a major source of cryptic chronic erythrocytic infections. The origin of such chronic infections is highly controversial as many key knowledge gaps remain unanswered. Yet, as parasites in these niches seem to be sheltered from immune response and antimalarial drugs, research on this area should be reinforced if elimination of malaria is to be achieved. Due to ethical and technical considerations, working with the liver, bone marrow and spleen from natural infections is very difficult. Recent advances in the development of humanized mouse models and organs-on-a-chip models, offer novel technological frontiers to study human diseases, vaccine validation and drug discovery. Here, we review current data of these frontier technologies in malaria, highlighting major challenges ahead to study P. vivax cryptic niches, which perpetuate transmission and burden.
Collapse
Affiliation(s)
- Iris Aparici Herraiz
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Hugo R. Caires
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Óscar Castillo-Fernández
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Núria Sima
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Lourdes Méndez-Mora
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
| | - Ruth M. Risueño
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Aurora Hernández-Machado
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
- Centre de Recerca Matemàtica (CRM), Barcelona, Spain
| | - Carmen Fernandez-Becerra
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Cristina C. Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Hernando A. del Portillo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- *Correspondence: Hernando A. del Portillo,
| |
Collapse
|
21
|
Wilder BK, Vigdorovich V, Carbonetti S, Minkah N, Hertoghs N, Raappana A, Cardamone H, Oliver BG, Trakhimets O, Kumar S, Dambrauskas N, Arredondo SA, Camargo N, Seilie AM, Murphy SC, Kappe SHI, Sather DN. Anti-TRAP/SSP2 monoclonal antibodies can inhibit sporozoite infection and may enhance protection of anti-CSP monoclonal antibodies. NPJ Vaccines 2022; 7:58. [PMID: 35618791 PMCID: PMC9135708 DOI: 10.1038/s41541-022-00480-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 04/22/2022] [Indexed: 11/10/2022] Open
Abstract
Vaccine-induced sterilizing protection from infection by Plasmodium parasites, the pathogens that cause malaria, will be essential in the fight against malaria as it would prevent both malaria-related disease and transmission. Stopping the relatively small number of parasites injected by the mosquito before they can migrate from the skin to the liver is an attractive means to this goal. Antibody-eliciting vaccines have been used to pursue this objective by targeting the major parasite surface protein present during this stage, the circumsporozoite protein (CSP). While CSP-based vaccines have recently had encouraging success in disease reduction, this was only achieved with extremely high antibody titers and appeared less effective for a complete block of infection (i.e., sterile protection). While such disease reduction is important, these and other results indicate that strategies focusing on CSP alone may not achieve the high levels of sterile protection needed for malaria eradication. Here, we show that monoclonal antibodies (mAbs) recognizing another sporozoite protein, TRAP/SSP2, exhibit a range of inhibitory activity and that these mAbs may augment CSP-based protection despite conferring no sterile protection on their own. Therefore, pursuing a multivalent subunit vaccine immunization is a promising strategy for improving infection-blocking malaria vaccines.
Collapse
Affiliation(s)
- Brandon K Wilder
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Vladimir Vigdorovich
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sara Carbonetti
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nana Minkah
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nina Hertoghs
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Andrew Raappana
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Hayley Cardamone
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Brian G Oliver
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Olesya Trakhimets
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nicholas Dambrauskas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Silvia A Arredondo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Annette M Seilie
- Department of Laboratory Medicine and Pathology and Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology and Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
22
|
Aleshnick M, Florez-Cuadros M, Martinson T, Wilder BK. Monoclonal antibodies for malaria prevention. Mol Ther 2022; 30:1810-1821. [PMID: 35395399 PMCID: PMC8979832 DOI: 10.1016/j.ymthe.2022.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 11/29/2022] Open
Abstract
Monoclonal antibodies are highly specific proteins that are cloned from a single B cell and bind to a single epitope on a pathogen. These laboratory-made molecules can serve as prophylactics or therapeutics for infectious diseases and have an impressive capacity to modulate the progression of disease, as demonstrated for the first time on a large scale during the COVID-19 pandemic. The high specificity and natural starting point of monoclonal antibodies afford an encouraging safety profile, yet the high cost of production remains a major limitation to their widespread use. While a monoclonal antibody approach to abrogating malaria infection is not yet available, the unique life cycle of the malaria parasite affords many opportunities for such proteins to act, and preliminary research into the efficacy of monoclonal antibodies in preventing malaria infection, disease, and transmission is encouraging. This review examines the current status and future outlook for monoclonal antibodies against malaria in the context of the complex life cycle and varied antigenic targets expressed in the human and mosquito hosts, and provides insight into the strengths and limitations of this approach to curtailing one of humanity’s oldest and deadliest diseases.
Collapse
Affiliation(s)
- Maya Aleshnick
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | - Thomas Martinson
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Brandon K Wilder
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA; Department of Parasitology, U.S. Naval Medical Research 6 (NAMRU-6), Lima, Peru
| |
Collapse
|
23
|
Goswami D, Kumar S, Betz W, Armstrong JM, Haile MT, Camargo N, Parthiban C, Seilie AM, Murphy SC, Vaughan AM, Kappe SH. A Plasmodium falciparum ATP binding cassette transporter is essential for liver stage entry into schizogony. iScience 2022; 25:104224. [PMID: 35521513 PMCID: PMC9061783 DOI: 10.1016/j.isci.2022.104224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/01/2022] [Accepted: 04/06/2022] [Indexed: 11/27/2022] Open
Abstract
Plasmodium sporozoites invade hepatocytes and transform into liver stages within a parasitophorous vacuole (PV). The parasites then grow and replicate their genome to form exoerythrocytic merozoites that infect red blood cells. We report that the human malaria parasite Plasmodium falciparum (Pf) expresses a C-type ATP-binding cassette transporter, Pf ABCC2, which marks the transition from invasive sporozoite to intrahepatocytic early liver stage. Using a humanized mouse infection model, we show that Pf ABCC2 localizes to the parasite plasma membrane in early and mid-liver stage parasites but is not detectable in late liver stages. Pf abcc2— sporozoites invade hepatocytes, form a PV, and transform into liver stage trophozoites but cannot transition to exoerythrocytic schizogony and fail to transition to blood stage infection. Thus, Pf ABCC2 is an expression marker for early phases of parasite liver infection and plays an essential role in the successful initiation of liver stage replication. Pf ABCC2 expression marks the transition from sporozoite to early liver stage Pf ABCC2 localizes to the early and mid-liver stage plasma membrane Pf ABCC2 is critical for initiation of exoerythrocytic schizogony Pf abcc2– liver stages fail to transition to blood stage infection
Collapse
|
24
|
Simwela NV, Waters AP. Current status of experimental models for the study of malaria. Parasitology 2022; 149:1-22. [PMID: 35357277 PMCID: PMC9378029 DOI: 10.1017/s0031182021002134] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Infection by malaria parasites (Plasmodium spp.) remains one of the leading causes of morbidity and mortality, especially in tropical regions of the world. Despite the availability of malaria control tools such as integrated vector management and effective therapeutics, these measures have been continuously undermined by the emergence of vector resistance to insecticides or parasite resistance to frontline antimalarial drugs. Whilst the recent pilot implementation of the RTS,S malaria vaccine is indeed a remarkable feat, highly effective vaccines against malaria remain elusive. The barriers to effective vaccines result from the complexity of both the malaria parasite lifecycle and the parasite as an organism itself with consequent major gaps in our understanding of their biology. Historically and due to the practical and ethical difficulties of working with human malaria infections, research into malaria parasite biology has been extensively facilitated by animal models. Animals have been used to study disease pathogenesis, host immune responses and their (dys)regulation and further disease processes such as transmission. Moreover, animal models remain at the forefront of pre-clinical evaluations of antimalarial drugs (drug efficacy, mode of action, mode of resistance) and vaccines. In this review, we discuss commonly used animal models of malaria, the parasite species used and their advantages and limitations which hinder their extrapolation to actual human disease. We also place into this context the most recent developments such as organoid technologies and humanized mice.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Andrew P. Waters
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| |
Collapse
|
25
|
Buchanan HD, Goodman CD, McFadden GI. Roles of the apicoplast across the life cycles of rodent and human malaria parasites. J Eukaryot Microbiol 2022; 69:e12947. [PMID: 36070203 PMCID: PMC9828729 DOI: 10.1111/jeu.12947] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Malaria parasites are diheteroxenous, requiring two hosts-a vertebrate and a mosquito-to complete their life cycle. Mosquitoes are the definitive host where malaria parasite sex occurs, and vertebrates are the intermediate host, supporting asexual amplification and more significant geographic spread. In this review, we examine the roles of a single malaria parasite compartment, the relict plastid known as the apicoplast, at each life cycle stage. We focus mainly on two malaria parasite species-Plasmodium falciparum and P. berghei-comparing the changing, yet ever crucial, roles of their apicoplasts.
Collapse
Affiliation(s)
- Hayley D. Buchanan
- Department of Infectious Diseases, Faculty of Medicine, Dentistry and Health Sciences, Melbourne Medical SchoolThe University of MelbourneMelbourneVic.Australia,Faculty of Science, School of BioSciencesThe University of MelbourneMelbourneVic.Australia
| | - Christopher D. Goodman
- Faculty of Science, School of BioSciencesThe University of MelbourneMelbourneVic.Australia
| | - Geoffrey I. McFadden
- Faculty of Science, School of BioSciencesThe University of MelbourneMelbourneVic.Australia
| |
Collapse
|
26
|
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: 74] [Impact Index Per Article: 18.5] [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
|
27
|
Wang LT, Pereira LS, Flores-Garcia Y, O'Connor J, Flynn BJ, Schön A, Hurlburt NK, Dillon M, Yang ASP, Fabra-García A, Idris AH, Mayer BT, Gerber MW, Gottardo R, Mason RD, Cavett N, Ballard RB, Kisalu NK, Molina-Cruz A, Nelson J, Vistein R, Barillas-Mury C, Amino R, Baker D, King NP, Sauerwein RW, Pancera M, Cockburn IA, Zavala F, Francica JR, Seder RA. A Potent Anti-Malarial Human Monoclonal Antibody Targets Circumsporozoite Protein Minor Repeats and Neutralizes Sporozoites in the Liver. Immunity 2020; 53:733-744.e8. [PMID: 32946741 PMCID: PMC7572793 DOI: 10.1016/j.immuni.2020.08.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/10/2020] [Accepted: 08/24/2020] [Indexed: 01/05/2023]
Abstract
Discovering potent human monoclonal antibodies (mAbs) targeting the Plasmodium falciparum circumsporozoite protein (PfCSP) on sporozoites (SPZ) and elucidating their mechanisms of neutralization will facilitate translation for passive prophylaxis and aid next-generation vaccine development. Here, we isolated a neutralizing human mAb, L9 that preferentially bound NVDP minor repeats of PfCSP with high affinity while cross-reacting with NANP major repeats. L9 was more potent than six published neutralizing human PfCSP mAbs at mediating protection against mosquito bite challenge in mice. Isothermal titration calorimetry and multiphoton microscopy showed that L9 and the other most protective mAbs bound PfCSP with two binding events and mediated protection by killing SPZ in the liver and by preventing their egress from sinusoids and traversal of hepatocytes. This study defines the subdominant PfCSP minor repeats as neutralizing epitopes, identifies an in vitro biophysical correlate of SPZ neutralization, and demonstrates that the liver is an important site for antibodies to prevent malaria.
Collapse
Affiliation(s)
- Lawrence T Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lais S Pereira
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - James O'Connor
- Department of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia; The Australian National University Medical School, Canberra, ACT 2601, Australia
| | - Barbara J Flynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Arne Schön
- Department of Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Nicholas K Hurlburt
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Marlon Dillon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Annie S P Yang
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Amanda Fabra-García
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Azza H Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bryan T Mayer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Monica W Gerber
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Rosemarie D Mason
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Cavett
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Reid B Ballard
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neville K Kisalu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alvaro Molina-Cruz
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Jorgen Nelson
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Rachel Vistein
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Rogerio Amino
- Unit of Malaria Infection and Immunity, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - David Baker
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Neil P King
- Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Robert W Sauerwein
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marie Pancera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Ian A Cockburn
- Department of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia
| | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Joseph R Francica
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
28
|
Schäfer C, Roobsoong W, Kangwanrangsan N, Bardelli M, Rawlinson TA, Dambrauskas N, Trakhimets O, Parthiban C, Goswami D, Reynolds LM, Kennedy SY, Flannery EL, Murphy SC, Sather DN, Draper SJ, Sattabongkot J, Mikolajczak SA, Kappe SHI. A Humanized Mouse Model for Plasmodium vivax to Test Interventions that Block Liver Stage to Blood Stage Transition and Blood Stage Infection. iScience 2020; 23:101381. [PMID: 32739836 PMCID: PMC7399188 DOI: 10.1016/j.isci.2020.101381] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/02/2020] [Accepted: 07/15/2020] [Indexed: 12/20/2022] Open
Abstract
The human malaria parasite Plasmodium vivax remains vastly understudied, mainly due to the lack of suitable laboratory models. Here, we report a humanized mouse model to test interventions that block P. vivax parasite transition from liver stage infection to blood stage infection. Human liver-chimeric FRGN huHep mice infected with P. vivax sporozoites were infused with human reticulocytes, allowing transition of exo-erythrocytic merozoites to reticulocyte infection and development into all erythrocytic forms, including gametocytes, in vivo. In order to test the utility of this model for preclinical assessment of interventions, the invasion blocking potential of a monoclonal antibody targeting the essential interaction of the P. vivax Duffy Binding Protein with the Duffy antigen receptor was tested by passive immunization. This antibody inhibited invasion by over 95%, providing unprecedented in vivo evidence that PvDBP constitutes a promising blood stage vaccine candidate and proving our model highly suitable to test blood stage interventions.
Collapse
Affiliation(s)
- Carola Schäfer
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Niwat Kangwanrangsan
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | | | - Nicholas Dambrauskas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Olesya Trakhimets
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Chaitra Parthiban
- Departments of Laboratory Medicine and Microbiology and Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Laura M Reynolds
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Spencer Y Kennedy
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Erika L Flannery
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Sean C Murphy
- Departments of Laboratory Medicine and Microbiology and Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Sebastian A Mikolajczak
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
29
|
Goswami D, Betz W, Locham NK, Parthiban C, Brager C, Schäfer C, Camargo N, Nguyen T, Kennedy SY, Murphy SC, Vaughan AM, Kappe SH. A replication-competent late liver stage-attenuated human malaria parasite. JCI Insight 2020; 5:135589. [PMID: 32484795 DOI: 10.1172/jci.insight.135589] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/21/2020] [Indexed: 01/06/2023] Open
Abstract
Whole-sporozoite vaccines engender sterilizing immunity against malaria in animal models and importantly, in humans. Gene editing allows for the removal of specific parasite genes, enabling generation of genetically attenuated parasite (GAP) strains for vaccination. Using rodent malaria parasites, we have previously shown that late liver stage-arresting replication-competent (LARC) GAPs confer superior protection when compared with early liver stage-arresting replication-deficient GAPs and radiation-attenuated sporozoites. However, generating a LARC GAP in the human malaria parasite Plasmodium falciparum (P. falciparum) has been challenging. Here, we report the generation and characterization of a likely unprecedented P. falciparum LARC GAP generated by targeted gene deletion of the Mei2 gene: P. falciparum mei2-. Robust exoerythrocytic schizogony with extensive cell growth and DNA replication was observed for P. falciparum mei2- liver stages in human liver-chimeric mice. However, P. falciparum mei2- liver stages failed to complete development and did not form infectious exoerythrocytic merozoites, thereby preventing their transition to asexual blood stage infection. Therefore, P. falciparum mei2- is a replication-competent, attenuated human malaria parasite strain with potentially increased potency, useful for vaccination to protect against P. falciparum malaria infection.
Collapse
Affiliation(s)
- Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - William Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Navin K Locham
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | | | - Carolyn Brager
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Carola Schäfer
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Thao Nguyen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Spencer Y Kennedy
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | | | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA.,Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Stefan Hi Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA.,Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
30
|
Miyazaki S, Yang ASP, Geurten FJA, Marin-Mogollon C, Miyazaki Y, Imai T, Kolli SK, Ramesar J, Chevalley-Maurel S, Salman AM, van Gemert GJA, van Waardenburg YM, Franke-Fayard B, Hill AVS, Sauerwein RW, Janse CJ, Khan SM. Generation of Novel Plasmodium falciparum NF135 and NF54 Lines Expressing Fluorescent Reporter Proteins Under the Control of Strong and Constitutive Promoters. Front Cell Infect Microbiol 2020; 10:270. [PMID: 32587831 PMCID: PMC7298075 DOI: 10.3389/fcimb.2020.00270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022] Open
Abstract
Transgenic reporter lines of malaria parasites that express fluorescent or luminescent proteins are valuable tools for drug and vaccine screening assays as well as to interrogate parasite gene function. Different Plasmodium falciparum (Pf ) reporter lines exist, however nearly all have been created in the African NF54/3D7 laboratory strain. Here we describe the generation of novel reporter lines, using CRISPR/Cas9 gene modification, both in the standard Pf NF54 background and in a recently described Cambodian P. falciparum NF135.C10 line. Sporozoites of this line show more effective hepatocyte invasion and enhanced liver merozoite development compared to Pf NF54. We first generated Pf NF54 reporter parasites to analyze two novel promoters for constitutive and high expression of mCherry-luciferase and GFP in blood and mosquito stages. The promoter sequences were selected based on available transcriptome data and are derived from two housekeeping genes, i.e., translation initiation factor SUI1, putative (sui1, PF3D7_1243600) and 40S ribosomal protein S30 (40s, PF3D7_0219200). We then generated and characterized reporter lines in the Pf NF135.C10 line which express GFP driven by the sui1 and 40s promoters as well as by the previously used ef1α promoter (GFP@ef1α, GFP@sui1, GFP@40s). The GFP@40s reporter line showed strongest GFP expression in liver stages as compared to the other two lines. The strength of reporter expression by the 40s promoter throughout the complete life cycle, including liver stages, makes transgenic lines expressing reporters by the 40s promoter valuable novel tools for analyses of P. falciparum.
Collapse
Affiliation(s)
- Shinya Miyazaki
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Annie S P Yang
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Fiona J A Geurten
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Yukiko Miyazaki
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Takashi Imai
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Surendra Kumar Kolli
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Jai Ramesar
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Ahmed M Salman
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Geert-Jan A van Gemert
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Youri M van Waardenburg
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Adrian V S Hill
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,TropIQ Health Sciences, Nijmegen, Netherlands
| | - Chris J Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Shahid M Khan
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
31
|
Ragotte RJ, Higgins MK, Draper SJ. The RH5-CyRPA-Ripr Complex as a Malaria Vaccine Target. Trends Parasitol 2020; 36:545-559. [PMID: 32359873 PMCID: PMC7246332 DOI: 10.1016/j.pt.2020.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 11/04/2022]
Abstract
Despite ongoing efforts, a highly effective vaccine against Plasmodium falciparum remains elusive. Vaccines targeting the pre-erythrocytic stages of the P. falciparum life cycle are the most advanced to date, affording moderate levels of efficacy in field trials. However, the discovery that the members of the merozoite PfRH5-PfCyRPA-PfRipr (RCR) complex are capable of inducing strain-transcendent neutralizing antibodies has renewed enthusiasm for the possibility of preventing disease by targeting the parasite during the blood stage of infection. With Phase I/II clinical trials now underway using first-generation vaccines against PfRH5, and more on the horizon for PfCyRPA and PfRipr, this review explores the rationale and future potential of the RCR complex as a P. falciparum vaccine target.
Collapse
Affiliation(s)
- Robert J Ragotte
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK.
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
32
|
Abstract
There is a pressing need for compounds with broad-spectrum activity against malaria parasites at various life cycle stages to achieve malaria elimination. However, this goal cannot be accomplished without targeting the tenacious dormant liver-stage hypnozoite that causes multiple relapses after the first episode of illness. In the search for the magic bullet to radically cure Plasmodium vivax malaria, tafenoquine outperformed other candidate drugs and was approved by the U.S. Food and Drug Administration in 2018. Tafenoquine is an 8-aminoquinoline that inhibits multiple life stages of various Plasmodium species. Additionally, its much longer half-life allows for single-dose treatment, which will improve the compliance rate. Despite its approval and the long-time use of other 8-aminoquinolines, the mechanisms behind tafenoquine's activity and adverse effects are still largely unknown. In this Perspective, we discuss the plausible underlying mechanisms of tafenoquine's antiparasitic activity and highlight its role as a cellular stressor. We also discuss potential drug combinations and the development of next-generation 8-aminoquinolines to further improve the therapeutic index of tafenoquine for malaria treatment and prevention.
Collapse
Affiliation(s)
- Kuan-Yi Lu
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, North Carolina 27708, United States
| | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, North Carolina 27708, United States.,Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
33
|
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: 100] [Impact Index Per Article: 16.7] [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
|
34
|
Goh YS, McGuire D, Rénia L. Vaccination With Sporozoites: Models and Correlates of Protection. Front Immunol 2019; 10:1227. [PMID: 31231377 PMCID: PMC6560154 DOI: 10.3389/fimmu.2019.01227] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Despite continuous efforts, the century-old goal of eradicating malaria still remains. Multiple control interventions need to be in place simultaneously to achieve this goal. In addition to effective control measures, drug therapies and insecticides, vaccines are critical to reduce mortality and morbidity. Hence, there are numerous studies investigating various malaria vaccine candidates. Most of the malaria vaccine candidates are subunit vaccines. However, they have shown limited efficacy in Phase II and III studies. To date, only whole parasite formulations have been shown to induce sterile immunity in human. In this article, we review and discuss the recent developments in vaccination with sporozoites and the mechanisms of protection involved.
Collapse
Affiliation(s)
- Yun Shan Goh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore
| | - Daniel McGuire
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
35
|
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: 6.2] [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
|
36
|
Marin-Mogollon C, Salman AM, Koolen KMJ, Bolscher JM, van Pul FJA, Miyazaki S, Imai T, Othman AS, Ramesar J, van Gemert GJ, Kroeze H, Chevalley-Maurel S, Franke-Fayard B, Sauerwein RW, Hill AVS, Dechering KJ, Janse CJ, Khan SM. A P. falciparum NF54 Reporter Line Expressing mCherry-Luciferase in Gametocytes, Sporozoites, and Liver-Stages. Front Cell Infect Microbiol 2019; 9:96. [PMID: 31058097 PMCID: PMC6477837 DOI: 10.3389/fcimb.2019.00096] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/22/2019] [Indexed: 12/16/2022] Open
Abstract
Transgenic malaria parasites expressing fluorescent and bioluminescent proteins are valuable tools to interrogate malaria-parasite biology and to evaluate drugs and vaccines. Using CRISPR/Cas9 methodology a transgenic Plasmodium falciparum (Pf) NF54 line was generated that expresses a fusion of mCherry and luciferase genes under the control of the Pf etramp10.3 gene promoter (line mCherry-luc@etramp10.3). Pf etramp10.3 is related to rodent Plasmodium uis4 and the uis4 promoter has been used to drive high transgene expression in rodent parasite sporozoites and liver-stages. We examined transgene expression throughout the complete life cycle and compared this expression to transgenic lines expressing mCherry-luciferase and GFP-luciferase under control of the constitutive gapdh and eef1a promoters. The mCherry-luc@etramp10.3 parasites express mCherry in gametocytes, sporozoites, and liver-stages. While no mCherry signal was detected in asexual blood-stage parasites above background levels, luciferase expression was detected in asexual blood-stages, as well as in gametocytes, sporozoites and liver-stages, with the highest levels of reporter expression detected in stage III-V gametocytes and in sporozoites. The expression of mCherry and luciferase in gametocytes and sporozoites makes this transgenic parasite line suitable to use in in vitro assays that examine the effect of transmission blocking inhibitors and to analyse gametocyte and sporozoite biology.
Collapse
Affiliation(s)
| | - Ahmed M Salman
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Fiona J A van Pul
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Shinya Miyazaki
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Takashi Imai
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ahmad Syibli Othman
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Jai Ramesar
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hans Kroeze
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Robert W Sauerwein
- TropIQ Health Sciences, Nijmegen, Netherlands.,Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Adrian V S Hill
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Chris J Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Shahid M Khan
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
37
|
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
|
38
|
Tyagi RK, Tandel N, Deshpande R, Engelman RW, Patel SD, Tyagi P. Humanized Mice Are Instrumental to the Study of Plasmodium falciparum Infection. Front Immunol 2018; 9:2550. [PMID: 30631319 PMCID: PMC6315153 DOI: 10.3389/fimmu.2018.02550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/17/2018] [Indexed: 02/05/2023] Open
Abstract
Research using humanized mice has advanced our knowledge and understanding of human haematopoiesis, non-adaptive and adaptive immunity, autoimmunity, infectious disease, cancer biology, and regenerative medicine. Challenges posed by the human-malaria parasite Plasmodium falciparum include its complex life cycle, the evolution of drug resistance against anti-malarials, poor diagnosis, and a lack of effective vaccines. Advancements in genetically engineered and immunodeficient mouse strains, have allowed for studies of the asexual blood stage, exoerythrocytic stage and the transition from liver-to-blood stage infection, in a single vertebrate host. This review discusses the process of "humanization" of various immunodeficient/transgenic strains and their contribution to translational biomedical research. Our work reviews the strategies employed to overcome the remaining-limitations of the developed human-mouse chimera(s).
Collapse
Affiliation(s)
- Rajeev K. Tyagi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Biomedical parasitology Unit, Institute Pasteur, Paris, France
- Department of Global Health, College of Public Health, University of South Florida, Tampa, FL, United States
| | - Nikunj Tandel
- Institute of Science, Nirma University, Ahmedabad, India
| | | | - Robert W. Engelman
- Department of Pediatrics, Pathology and Cell Biology, University of South Florida, Tampa, FL, United States
| | | | - Priyanka Tyagi
- Department of Basic and Applied Sciences, School of Engineering, GD Goenka University, Gurgaon, India
| |
Collapse
|
39
|
Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat Immunol 2018; 19:1199-1211. [PMID: 30333613 DOI: 10.1038/s41590-018-0228-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/31/2018] [Indexed: 02/08/2023]
Abstract
Development of a malaria vaccine remains a critical priority to decrease clinical disease and mortality and facilitate eradication. Accordingly, RTS,S, a protein-subunit vaccine, has completed phase III clinical trials and confers ~30% protection against clinical infection over 4 years. Whole-attenuated-sporozoite and viral-subunit vaccines induce between 20% and 100% protection against controlled human malaria infection, but there is limited published evidence to date for durable, high-level efficacy (>50%) against natural exposure. Importantly, fundamental scientific advances related to the potency, durability, breadth and location of immune responses will be required for improving vaccine efficacy with these and other vaccine approaches. In this Review, we focus on the current understanding of immunological mechanisms of protection from animal models and human vaccine studies, and on how these data should inform the development of next-generation vaccines. Furthermore, we introduce the concept of using passive immunization with monoclonal antibodies as a new approach to prevent and eliminate malaria.
Collapse
Affiliation(s)
- Ian A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
40
|
Glennon EKK, Dankwa S, Smith JD, Kaushansky A. Opportunities for Host-targeted Therapies for Malaria. Trends Parasitol 2018; 34:843-860. [PMID: 30122551 PMCID: PMC6168423 DOI: 10.1016/j.pt.2018.07.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 12/19/2022]
Abstract
Despite the recent successes of artemisinin-based antimalarial drugs, many still die from severe malaria, and eradication efforts are hindered by the limited drugs currently available to target transmissible gametocyte parasites and liver-resident dormant Plasmodium vivax hypnozoites. Host-targeted therapy is a new direction for infectious disease drug development and aims to interfere with host molecules, pathways, or networks that are required for infection or that contribute to disease. Recent advances in our understanding of host pathways involved in parasite development and pathogenic mechanisms in severe malaria could facilitate the development of host-targeted interventions against Plasmodium infection and malaria disease. This review discusses new opportunities for host-targeted therapeutics for malaria and the potential to harness drug polypharmacology to simultaneously target multiple host pathways using a single drug intervention.
Collapse
Affiliation(s)
- Elizabeth K K Glennon
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory, Box 357965, Seattle, WA 98195, USA; These authors made an equal contribution
| | - Selasi Dankwa
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; These authors made an equal contribution
| | - Joseph D Smith
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory, Box 357965, Seattle, WA 98195, USA
| | - Alexis Kaushansky
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory, Box 357965, Seattle, WA 98195, USA.
| |
Collapse
|
41
|
Minkah NK, Schafer C, Kappe SHI. Humanized Mouse Models for the Study of Human Malaria Parasite Biology, Pathogenesis, and Immunity. Front Immunol 2018; 9:807. [PMID: 29725334 PMCID: PMC5917005 DOI: 10.3389/fimmu.2018.00807] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/03/2018] [Indexed: 12/25/2022] Open
Abstract
Malaria parasite infection continues to inflict extensive morbidity and mortality in resource-poor countries. The insufficiently understood parasite biology, continuously evolving drug resistance and the lack of an effective vaccine necessitate intensive research on human malaria parasites that can inform the development of new intervention tools. Humanized mouse models have been greatly improved over the last decade and enable the direct study of human malaria parasites in vivo in the laboratory. Nevertheless, no small animal model developed so far is capable of maintaining the complete life cycle of Plasmodium parasites that infect humans. The ultimate goal is to develop humanized mouse systems in which a Plasmodium infection closely reproduces all stages of a parasite infection in humans, including pre-erythrocytic infection, blood stage infection and its associated pathology, transmission as well as the human immune response to infection. Here, we discuss current humanized mouse models and the future directions that should be taken to develop next-generation models for human malaria parasite research.
Collapse
Affiliation(s)
- Nana K Minkah
- Center for Infectious Disease Research, Seattle, WA, United States
| | - Carola Schafer
- Center for Infectious Disease Research, Seattle, WA, United States
| | - Stefan H I Kappe
- Center for Infectious Disease Research, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|