1
|
Deshmukh B, Khatri D, Kochar SK, Athale C, Karmodiya K. In vitro evaluation of multi-protein chimeric antigens in effectively clearing the blood stage of Plasmodium falciparum. Vaccine 2025; 53:126952. [PMID: 40037124 DOI: 10.1016/j.vaccine.2025.126952] [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: 10/11/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/06/2025]
Abstract
Plasmodium falciparum-induced malaria remains a fatal disease affecting millions of people worldwide. Mainly, the blood stage of malaria is highly pathogenic and symptomatic, rapidly damaging the host organs and occasionally leading to death. Currently, no vaccines are approved for use against the blood stage of malaria. Canonical vaccines in the past have selected the most immunodominant or essential protein to block the growth of the parasite. This strategy works efficiently for low-complexity organisms such as viruses and a few bacteria but has not shown promising results for a malaria vaccine. Plasmodium has a complex life cycle and vaccine candidates especially during blood stage are ineffective due to multiple gene families showing redundancy, immune evasion, and insufficient antibody titer. Herein, we demonstrate a strategy of combining multiple antigens from the blood stage of Plasmodium falciparum using only the most immunodominant peptide sequences as a way of tackling polymorphism and redundancy. We created three chimeric antigens targeting eight PfEMP1 proteins (chimeric varB) and eight merozoite surface proteins (chimeric MSP and InvP) by selecting and stitching B-cell epitopes. Our chimeric constructs show naturally circulating antibodies against individual peptides using epitope-mapping microarray as well as entire proteins in malaria-infected patients. We demonstrate that anti-varB antibodies are neutralizing in nature and significantly reduce the cytoadhesion on an organ-on-chip system with a microfluidic device mimicking physiological conditions. We have applied a Deep Learning based method to quantify the number of adhered RBCs under fluidic conditions that is used to study cytoadhesion. Furthermore, the anti-MSP and InvP antibodies show complete growth inhibition in a single cycle at a combined concentration of 0.13 mg/ml. Overall, our preliminary results show that a combination of antigenic peptides from multiple antigens can potentially effectively reduce cytoadhesion and clear blood stage infection in in-vitro settings.
Collapse
Affiliation(s)
- Bhagyashree Deshmukh
- Indian Institute of Science Education and Research, Pashan, Pune, Maharashtra 411008, India
| | - Dhruv Khatri
- Indian Institute of Science Education and Research, Pashan, Pune, Maharashtra 411008, India
| | | | - Chaitanya Athale
- Indian Institute of Science Education and Research, Pashan, Pune, Maharashtra 411008, India
| | - Krishanpal Karmodiya
- Indian Institute of Science Education and Research, Pashan, Pune, Maharashtra 411008, India.
| |
Collapse
|
2
|
Hamisi MA, Asri NAM, Yassim ASM, Suppian R. A systematic review on malaria and Tuberculosis (TB) vaccine challenges in sub-Saharan African clinical trials. PLoS One 2025; 20:e0317233. [PMID: 39854359 PMCID: PMC11760592 DOI: 10.1371/journal.pone.0317233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 12/25/2024] [Indexed: 01/26/2025] Open
Abstract
OBJECTIVE For more than a century, developing novel and effective vaccines against malaria and Tuberculosis (TB) infections has been a challenge. This review sought to investigate the reasons for the slow progress of malaria and TB vaccine candidates in sub-Saharan African clinical trials. METHODS The systematic review protocol was registered on PROSPERO on July 26, 2023 (CRD42023445166). The research articles related to the immunogenicity, efficacy, or safety of malaria or TB vaccines that were published between January 1, 2012, and August 31, 2023, were searched on three databases: Web of Science (WoS), PubMed, and ClinicalTrials.gov. RESULTS A total of 2342 articles were obtained, 50 of which met the inclusion criteria. 28 (56%) articles reported on malaria vaccine attributes, while 22 (44%) articles reported on TB vaccines. In both cases, the major challenges in sub-Saharan African clinical trials were immunogenicity and efficacy, rather than safety. CONCLUSION Factors such as population characteristics, pathogen genetic diversity, vaccine nature, strategy, and formulation were associated with slow progress of the malaria and TB vaccine candidates in sub-Saharan African clinical trials.
Collapse
Affiliation(s)
- Maonezi Abas Hamisi
- School of Science and Technical Education (CoSTE), Mbeya University of Science and Technology, Mbeya, Tanzania
| | - Nur Ain Mohd Asri
- School of Health Sciences, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| | | | - Rapeah Suppian
- School of Health Sciences, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
3
|
Weth AF, Dangerfield EM, Timmer MSM, Stocker BL. Recent Advances in the Development of Mincle-Targeting Vaccine Adjuvants. Vaccines (Basel) 2024; 12:1320. [PMID: 39771982 PMCID: PMC11680293 DOI: 10.3390/vaccines12121320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 01/11/2025] Open
Abstract
The Macrophage-inducible C-type lectin (Mincle) is a pattern-recognition receptor (PRR), which has shown much promise as a molecular target for the development of TH1/TH17-skewing vaccine adjuvants. In 2009, the first non-proteinaceous Mincle ligands, trehalose dimycolate (TDM) and trehalose dibehenate (TDB), were identified. This prompted a search for other Mincle agonists and the exploration of Mincle agonists as vaccine adjuvants for both preventative and therapeutic (anti-cancer) vaccines. In this review, we discuss those classes of Mincle agonists that have been explored for their adjuvant potential. These Mincle agonists have been used as stand-alone adjuvants or in combination with other pathogen-associated molecular patterns (PAMPs) or immunomodulatory agents. We will also highlight recently identified Mincle ligands with hitherto unknown adjuvanticity. Conjugate vaccines that contain covalently linked adjuvants and/or adjuvant-antigen combinations are also presented, as well as the different formulations (e.g., oil-in-water emulsions, liposomes, and particulate delivery systems) that have been used for the codelivery of antigens and adjuvants. Insofar the reader is presented with a thorough review of the potential of Mincle-mediated vaccine adjuvants, including historical context, present-day research and clinical trials, and outstanding research questions, such as the role of ligand presentation and Mincle clustering, which, if better understood, will aid in the development of the much-needed TH1/TH17-skewing vaccine adjuvants.
Collapse
Affiliation(s)
| | | | - Mattie S. M. Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Bridget L. Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| |
Collapse
|
4
|
Chutiyami M, Saravanakumar P, Bello UM, Salihu D, Adeleye K, Kolo MA, Dawa KK, Hamina D, Bhandari P, Sulaiman SK, Sim J. Malaria vaccine efficacy, safety, and community perception in Africa: a scoping review of recent empirical studies. Infection 2024; 52:2007-2028. [PMID: 38441731 PMCID: PMC11499420 DOI: 10.1007/s15010-024-02196-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/22/2024] [Indexed: 10/24/2024]
Abstract
AIM The review summarizes the recent empirical evidence on the efficacy, safety, and community perception of malaria vaccines in Africa. METHODS Academic Search Complete, African Journals Online, CINAHL, Medline, PsychInfo, and two gray literature sources were searched in January 2023, and updated in June 2023. Relevant studies published from 2012 were included. Studies were screened, appraised, and synthesized in line with the review aim. Statistical results are presented as 95% Confidence Intervals and proportions/percentages. RESULTS Sixty-six (N = 66) studies met the inclusion criteria. Of the vaccines identified, overall efficacy at 12 months was highest for the R21 vaccine (N = 3) at 77.0%, compared to the RTS,S vaccine (N = 15) at 55%. The efficacy of other vaccines was BK-SE36 (11.0-50.0%, N = 1), ChAd63/MVA ME-TRAP (- 4.7-19.4%, N = 2), FMP2.1/AS02A (7.6-9.9%, N = 1), GMZ2 (0.6-60.0%, N = 5), PfPZ (20.0-100.0%, N = 5), and PfSPZ-CVac (24.8-33.6%, N = 1). Injection site pain and fever were the most common adverse events (N = 26), while febrile convulsion (N = 8) was the most reported, vaccine-related Serious Adverse Event. Mixed perceptions of malaria vaccines were found in African communities (N = 17); awareness was generally low, ranging from 11% in Tanzania to 60% in Nigeria (N = 9), compared to willingness to accept the vaccines, which varied from 32.3% in Ethiopia to 96% in Sierra Leone (N = 15). Other issues include availability, logistics, and misconceptions. CONCLUSION Malaria vaccines protect against malaria infection in varying degrees, with severe side effects rarely occurring. Further research is required to improve vaccine efficacy and community involvement is needed to ensure successful widespread use in African communities.
Collapse
Affiliation(s)
- Muhammad Chutiyami
- School of Nursing and Midwifery, University of Technology Sydney, Sydney, Australia.
| | - Priya Saravanakumar
- School of Nursing and Midwifery, University of Technology Sydney, Sydney, Australia
| | - Umar Muhammad Bello
- Department of Physiotherapy and Paramedicine, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Dauda Salihu
- College of Nursing, Jouf University, Sakaka, Saudi Arabia
| | - Khadijat Adeleye
- College of Nursing, University of Massachusetts, Amherst, MA, 01003, USA
| | | | - Kabiru Kasamu Dawa
- School of Nursing, Midwifery and Health Education, University of Bedfordshire, Luton, UK
| | - Dathini Hamina
- Department of Nursing Science, University of Maiduguri, Maiduguri, Nigeria
| | - Pratibha Bhandari
- School of Nursing and Midwifery, University of Technology Sydney, Sydney, Australia
| | | | - Jenny Sim
- WHO Collaborating Centre for Nursing, Midwifery and Health Development, University of Technology Sydney, Sydney, Australia
- School of Nursing, Midwifery and Paramedicine, Australian Catholic University, Sydney, Australia
| |
Collapse
|
5
|
Boyle MJ, Engwerda CR, Jagannathan P. The impact of Plasmodium-driven immunoregulatory networks on immunity to malaria. Nat Rev Immunol 2024; 24:637-653. [PMID: 38862638 PMCID: PMC11688169 DOI: 10.1038/s41577-024-01041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 06/13/2024]
Abstract
Malaria, caused by infection with Plasmodium parasites, drives multiple regulatory responses across the immune landscape. These regulatory responses help to protect against inflammatory disease but may in some situations hamper the acquisition of adaptive immune responses that clear parasites. In addition, the regulatory responses that occur during Plasmodium infection may negatively affect malaria vaccine efficacy in the most at-risk populations. Here, we discuss the specific cellular mechanisms of immunoregulatory networks that develop during malaria, with a focus on knowledge gained from human studies and studies that involve the main malaria parasite to affect humans, Plasmodium falciparum. Leveraging this knowledge may lead to the development of new therapeutic approaches to increase protective immunity to malaria during infection or after vaccination.
Collapse
Affiliation(s)
- Michelle J Boyle
- Life Sciences Division, Burnet Institute, Melbourne, Victoria, Australia.
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| | | | - Prasanna Jagannathan
- Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
6
|
Li J, Docile HJ, Fisher D, Pronyuk K, Zhao L. Current Status of Malaria Control and Elimination in Africa: Epidemiology, Diagnosis, Treatment, Progress and Challenges. J Epidemiol Glob Health 2024; 14:561-579. [PMID: 38656731 PMCID: PMC11442732 DOI: 10.1007/s44197-024-00228-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
The African continent carries the greatest malaria burden in the world. Falciparum malaria especially has long been the leading cause of death in Africa. Climate, economic factors, geographical location, human intervention and unstable security are factors influencing malaria transmission. Due to repeated infections and early interventions, the proportion of clinically atypical malaria or asymptomatic plasmodium carriers has increased significantly, which easily lead to misdiagnosis and missed diagnosis. African countries have made certain progress in malaria control and elimination, including rapid diagnosis of malaria, promotion of mosquito nets and insecticides, intermittent prophylactic treatment in high-risk groups, artemisinin based combination therapies, and the development of vaccines. Between 2000 and 2022, there has been a 40% decrease in malaria incidence and a 60% reduction in mortality rate in the WHO African Region. However, many challenges are emerging in the fight against malaria in Africa, such as climate change, poverty, substandard health services and coverage, increased outdoor transmission and the emergence of new vectors, and the growing threat of resistance to antimalarial drugs and insecticides. Joint prevention and treatment, identifying molecular determinants of resistance, new drug development, expanding seasonal malaria chemo-prevention intervention population, and promoting the vaccination of RTS, S/AS01 and R21/Matrix-M may help to solve the dilemma. China's experience in eliminating malaria is conducive to Africa's malaria prevention and control, and China-Africa cooperation needs to be constantly deepened and advanced. Our review aims to help the global public develop a comprehensive understanding of malaria in Africa, thereby contributing to malaria control and elimination.
Collapse
Affiliation(s)
- Jiahuan Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Haragakiza Jean Docile
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of The Western Cape, Cape Town, South Africa
| | - Khrystyna Pronyuk
- Department of Infectious Diseases, O. Bogomolets National Medical University, Kyiv, Ukraine
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
7
|
Wang R, Oliveira LVN, Hester MM, Carlson D, Christensen D, Specht CA, Levitz SM. Protection against experimental cryptococcosis elicited by Cationic Adjuvant Formulation 01-adjuvanted subunit vaccines. PLoS Pathog 2024; 20:e1012220. [PMID: 38976694 PMCID: PMC11257399 DOI: 10.1371/journal.ppat.1012220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/18/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024] Open
Abstract
The fungal infection, cryptococcosis, is responsible for >100,000 deaths annually. No licensed vaccines are available. We explored the efficacy and immune responses of subunit cryptococcal vaccines adjuvanted with Cationic Adjuvant Formulation 01 (CAF01). CAF01 promotes humoral and T helper (Th) 1 and Th17 immune responses and has been safely used in human vaccine trials. Four subcutaneous vaccines, each containing single recombinant Cryptococcus neoformans protein antigens, partially protected mice from experimental cryptococcosis. Protection increased, up to 100%, in mice that received bivalent and quadrivalent vaccine formulations. Vaccinated mice that received a pulmonary challenge with C. neoformans had an influx of leukocytes into the lung including robust numbers of polyfunctional CD4+ T cells which produced interferon gamma (IFNγ), tumor necrosis factor alpha (TNFα), and interleukin (IL)-17 upon ex vivo antigenic stimulation. Cytokine-producing lung CD8+ T cells were also found, albeit in lesser numbers. A significant, durable IFNγ response was observed in the lungs, spleen, and blood. Moreover, IFNγ secretion following ex vivo stimulation directly correlated with fungal control in the lungs. Thus, we have developed multivalent cryptococcal vaccines which protect mice from experimental cryptococcosis using an adjuvant which has been safely tested in humans. These preclinical studies suggest a path towards human cryptococcal vaccine trials.
Collapse
Affiliation(s)
- Ruiying Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Lorena V. N. Oliveira
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Maureen M. Hester
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Diana Carlson
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | | | - Charles A. Specht
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Stuart M. Levitz
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
8
|
Šťastná E, Erbs G, Skovgaard K, Jakobsen JT, Bailey M, Pedersen GK, Jungersen G. Effects of different immunomodulating liposome-based adjuvants and injection sites on immunogenicity in pigs. Microbes Infect 2024; 26:105346. [PMID: 38670217 DOI: 10.1016/j.micinf.2024.105346] [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: 02/13/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
Vaccine adjuvants, such as liposome-based cationic adjuvant formulations (CAFs), are able to boost immune responses and, by incorporation of distinct immunomodulators, steer immunity towards a desired direction in mice, non-human primates and humans, while less studied in pigs. Here we used commercial pigs to investigate polarizing adjuvant effects of CAFs with immunomodulators: C-type lectin receptor ligands trehalose-6,6'-dibehenate and monomycolyl glycerol, toll-like receptor 3 ligand Poly(I:C) or retinoic acid. Vaccines were formulated with a recombinant Chlamydia model protein antigen and administered via three injection routes. All adjuvants significantly increased antigen-specific IgG in serum, compared to non-adjuvanted antigen. Administering the vaccines through intramuscular and intraperitoneal routes induced significantly higher antigen-specific IgG and IgA serum antibodies, than the perirectal route. Although immunizations triggered cell-mediated immunity, no significant differences between adjuvants or injection sites were detected. Genes depicting T cell subtypes revealed only minor differences. Our findings suggest that specific signatures of the tested adjuvant immunomodulation do not translate well from mice to pigs in standard two-dose immunizations. This study provides new insights into immune responses to CAFs in pigs, and highlights that adjuvant development should ideally be carried out in the intended species of interest or in models with high predictive validity/translational value.
Collapse
Affiliation(s)
- Evelína Šťastná
- Infectious Disease Immunology, Centre for Vaccine Research, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Gitte Erbs
- Infectious Disease Immunology, Centre for Vaccine Research, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, 2800 Kongens Lyngby, Denmark
| | - Jeanne Toft Jakobsen
- Infectious Disease Immunology, Centre for Vaccine Research, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Mick Bailey
- Bristol Veterinary School, Langford House, University of Bristol, United Kingdom
| | - Gabriel Kristian Pedersen
- Infectious Disease Immunology, Centre for Vaccine Research, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Gregers Jungersen
- Infectious Disease Immunology, Centre for Vaccine Research, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark.
| |
Collapse
|
9
|
Wang R, Oliveira LVN, Hester MM, Carlson D, Christensen D, Specht CA, Levitz SM. Protection against experimental cryptococcosis elicited by Cationic Adjuvant Formulation 01-adjuvanted subunit vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.591045. [PMID: 38712080 PMCID: PMC11071535 DOI: 10.1101/2024.04.24.591045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The fungal infection, cryptococcosis, is responsible for >100,000 deaths annually. No licensed vaccines are available. We explored the efficacy and immune responses of subunit cryptococcal vaccines adjuvanted with Cationic Adjuvant Formulation 01 (CAF01). CAF01 promotes humoral and T helper (Th) 1 and Th17 immune responses and has been safely used in human vaccine trials. Four subcutaneous vaccines, each containing single recombinant Cryptococcus neoformans protein antigens, partially protected mice from experimental cryptococcosis. Protection increased, up to 100%, in mice that received bivalent and quadrivalent vaccine formulations. Vaccinated mice that received a pulmonary challenge with C. neoformans had an influx of leukocytes into the lung including robust numbers of polyfunctional CD4+ T cells which produced Interferon gamma (IFNγ), tumor necrosis factor alpha (TNFα), and interleukin (IL)-17 upon ex vivo antigenic stimulation. Cytokine-producing lung CD8+ T cells were also found, albeit in lesser numbers. A significant, durable IFNγ response was observed in the lungs, spleen, and blood. Moreover, IFNγ secretion following ex vivo stimulation directly correlated with fungal clearance in the lungs. Thus, we have developed multivalent cryptococcal vaccines which protect mice from experimental cryptococcosis using an adjuvant which has been safely tested in humans. These preclinical studies suggest a path towards human cryptococcal vaccine trials.
Collapse
Affiliation(s)
- Ruiying Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lorena V. N. Oliveira
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Maureen M. Hester
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Diana Carlson
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dennis Christensen
- Statens Serum Institut, Copenhagen, Denmark
- Present address: Croda Pharma, Diplomvej 381, Lyngby 2800, Denmark
| | - Charles A. Specht
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Stuart M. Levitz
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
10
|
Ogwang R, Murugu L, Nkumama IN, Nyamako L, Kai O, Mwai K, Murungi L, Idro R, Bejon P, Tuju J, Kinyanjui SM, Osier FHA. Bi-isotype immunoglobulins enhance antibody-mediated neutrophil activity against Plasmodium falciparum parasites. Front Immunol 2024; 15:1360220. [PMID: 38650925 PMCID: PMC11033408 DOI: 10.3389/fimmu.2024.1360220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
Background Malaria remains a major global health priority, and monoclonal antibodies (mAbs) are emerging as potential new tools to support efforts to control the disease. Recent data suggest that Fc-dependent mechanisms of immunity are important mediators of protection against the blood stages of the infection, but few studies have investigated this in the context of mAbs. We aimed to isolate mAbs agnostic to cognate antigens that target whole merozoites and simultaneously induce potent neutrophil activity measured by the level of reactive oxygen species (ROS) production using an antibody-dependent respiratory burst (ADRB) assay. Methods We used samples from semi-immune adults living in coastal Kenya to isolate mAbs that induce merozoite-specific ADRB activity. We then tested whether modifying the expressed IgG1 isotype to an IgG-IgA Fc region chimera would enhance the level of ADRB activity. Results We isolated a panel of nine mAbs with specificity to whole merozoites. mAb J31 induced ADRB activity in a dose-dependent fashion. Compared to IgG1, our modified antibody IgG-IgA bi-isotype induced higher ADRB activity across all concentrations tested. Further, we observed a negative hook effect at high IgG1 mAb concentrations (i.e., >200 µg/mL), but this was reversed by Fc modification. We identified MSP3.5 as the potential cognate target of mAb J31. Conclusions We demonstrate an approach to engineer mAbs with enhanced ADRB potency against blood-stage parasites.
Collapse
Affiliation(s)
- Rodney Ogwang
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- College of Health Sciences, Makerere University, Kampala, Uganda
| | - Lewis Murugu
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Biological Sciences, Pwani University, Kilifi, Kenya
| | - Irene N. Nkumama
- Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Lydia Nyamako
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Oscar Kai
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kennedy Mwai
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Linda Murungi
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Richard Idro
- College of Health Sciences, Makerere University, Kampala, Uganda
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Philip Bejon
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - James Tuju
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Biological Sciences, Pwani University, Kilifi, Kenya
| | - Sam Muchina Kinyanjui
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Faith H. A. Osier
- Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
11
|
Plieskatt J, Bang P, Wood GK, Naghizadeh M, Singh SK, Jore MM, Theisen M. Clinical formulation development of Plasmodium falciparum malaria vaccine candidates based on Pfs48/45, Pfs230, and PfCSP. Vaccine 2024; 42:1980-1992. [PMID: 38388238 DOI: 10.1016/j.vaccine.2024.02.043] [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: 10/09/2023] [Revised: 01/09/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Two malaria transmission-blocking vaccine (TBV) candidates, R0.6C and ProC6C, have completed preclinical development including the selection of adjuvants, Alhydrogel® with or without the saponin based adjuvant Matrix-M™. Here, we report on the final drug product (formulation) design of R0.6C and ProC6C and evaluate their safety and biochemical stability in preparation for preclinical and clinical pharmacy handling. The point-of-injection stability studies demonstrated that both the R0.6C and ProC6C antigens are stable on Alhydrogel in the presence or absence of Matrix-M for up to 24 h at room temperature. As this is the first study to combine Alhydrogel and Matrix-M for clinical use, we also evaluated their potential interactions. Matrix-M adsorbs to Alhydrogel, while not displacing the > 95 % adsorbed protein. The R0.6C and ProC6C formulations were found to be safe and well tolerated in repeated dose toxicity studies in rabbits generating high levels of functional antibodies that blocked infection of mosquitoes. Further, the R0.6C and ProC6C drug products were found to be stable for minimally 24 months when stored at 2-8 °C, with studies ongoing through 36 months. Together, this data demonstrates the safety and suitability of the L. lactis expression system as well as supports the clinical testing of the R0.6C and ProC6C malaria vaccine candidates in First-In-Human clinical trials.
Collapse
Affiliation(s)
- Jordan Plieskatt
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Bang
- Department of Vaccine Development, Statens Serum Institut, Copenhagen, Denmark
| | - Grith Krøyer Wood
- Department of Vaccine Development, Statens Serum Institut, Copenhagen, Denmark
| | - Mohammad Naghizadeh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Susheel K Singh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
Tomás-Cortázar J, Quinn C, Corcoran N, Blanco A, Christensen D, McClean S. BpOmpW antigen administered with CAF01 adjuvant stimulates comparable T cell responses to Sigma adjuvant system. Vaccine X 2024; 17:100438. [PMID: 38303776 PMCID: PMC10831100 DOI: 10.1016/j.jvacx.2024.100438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
There are no licensed vaccines to protect vulnerable populations from the potentially fatal tropical infection, melioidosis, despite its causative agent, Burkholderia pseudomallei, being endemic in tropical and subtropical regions. A promising vaccine candidate, BpOmpW protected mice from melioidosis infection for up to 81 days and stimulated robust interferon gamma responses in CD4+, CD8+, NK and NKT cells. In order to progress to human studies, selection of an adjuvant with an acceptable human safety profile that stimulates appropriate correlates of protection is essential. Here we demonstrate that the CAF01 vaccine adjuvant elicits optimal immune correlates of protection when administered with our BpOmpW vaccine. Specifically, we demonstrate that CAF01 administered with BpOmpW elicits robust Th1 responses, with potent IFN-γ responses in CD4+ and CD8+ T cells and NKT cells, in addition to Th17 and Th2 responses. This formulation will be particularly effective in protecting susceptible populations including people with type 2 diabetes from melioidosis.
Collapse
Affiliation(s)
- Julen Tomás-Cortázar
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Conor Quinn
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Niamh Corcoran
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Alfonso Blanco
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen S, Denmark
| | - Siobhán McClean
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
13
|
Poston TB. Advances in vaccine development for Chlamydia trachomatis. Pathog Dis 2024; 82:ftae017. [PMID: 39043447 PMCID: PMC11338180 DOI: 10.1093/femspd/ftae017] [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: 04/25/2024] [Revised: 06/18/2024] [Accepted: 07/25/2024] [Indexed: 07/25/2024] Open
Abstract
Chlamydia trachomatis is the most prevalent bacterial sexually transmitted infection globally. Antibiotic treatment is highly effective, but infection is often asymptomatic resulting in most individuals going undetected and untreated. This untreated infection can ascend to the upper female genital tract to cause pelvic inflammatory disease, tubal factor infertility, and ectopic pregnancy. Chlamydia screening and treatment programs have failed to control this epidemic and demonstrate the need for an efficacious vaccine to prevent transmission and disease. Animal models and human epidemiological data reveal that natural immunity can provide partial or short-lived sterilizing immunity. These data further demonstrate the importance of eliciting interferon gamma (IFNγ)-producing cluster of differentiation 4 (CD4) T cells (Th1 and Th1/17 cells) that can likely synergize with antibody-mediated opsonophagocytosis to provide optimal protection. These studies have guided preclinical rational vaccine design for decades and the first Phase 1 clinical trials have recently been completed. Recent advances have led to improvements in vaccine platforms and clinically safe adjuvants that help provide a path forward. This review describes vaccine models, correlates of immunity, antigen and adjuvant selection, and future clinical testing for Chlamydia vaccine development.
Collapse
Affiliation(s)
- Taylor B Poston
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| |
Collapse
|
14
|
Jamrozik E, Littler K, Meln I, Van Molle W, Morel S, Olesen OF, Rubbrecht M, Balasingam S, Neels P. Ethical approval for controlled human infectious model clinical trial protocols - A workshop report. Biologicals 2024; 85:101748. [PMID: 38350349 PMCID: PMC11004724 DOI: 10.1016/j.biologicals.2024.101748] [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: 12/06/2023] [Accepted: 01/28/2024] [Indexed: 02/15/2024] Open
Abstract
Controlled Human Infectious Model studies (CHIM) involve deliberately exposing volunteers to pathogens. To discuss ethical issues related to CHIM, the European Vaccine Initiative and the International Alliance for Biological Standardization organised the workshop "Ethical Approval for CHIM Clinical Trial Protocols", which took place on May 30-31, 2023, in Brussels, Belgium. The event allowed CHIM researchers, regulators, ethics committee (EC) members, and ethicists to examine the ethical criteria for CHIM and the role(s) of CHIM in pharmaceutical development. The discussions led to several recommendations, including continued assurance that routine ethical requirements are met, assurance that participants are well-informed, and that preparation of study documents must be both ethically and scientifically sound from an early stage. Study applications must clearly state the rationale for the challenge compared to alternative study designs. ECs need to have clear guidance and procedures for evaluating social value and assessing third-party risks. Among other things, public trust in research requires minimisation of harm to healthy volunteers and third-party risk. Other important considerations include appropriate stakeholder engagement, public education, and access to health care for participants after the study.
Collapse
Affiliation(s)
- Euzebiusz Jamrozik
- The Ethox Centre & Wellcome Centre for Ethics and Humanities, University of Oxford, Oxford, United Kingdom
| | - Katherine Littler
- Health Ethics and Governance Unit, Research for Health Department, Science Division, World Health Organization, Geneva, Switzerland
| | - Irina Meln
- European Vaccine Initiative, Heidelberg, Germany
| | | | | | - Ole F Olesen
- European Vaccine Initiative, Heidelberg, Germany
| | | | | | - Pieter Neels
- International Alliance for Biological Standardization (IABS), Lyon, France.
| |
Collapse
|
15
|
Martinez FJ, White M, Guillotte-Blisnick M, Huon C, Boucharlat A, Agou F, England P, Popovici J, Hou MM, Silk SE, Barrett JR, Nielsen CM, Reimer JM, Mukherjee P, Chauhan VS, Minassian AM, Draper SJ, Chitnis CE. PvDBPII elicits multiple antibody-mediated mechanisms that reduce growth in a Plasmodium vivax challenge trial. NPJ Vaccines 2024; 9:10. [PMID: 38184681 PMCID: PMC10771494 DOI: 10.1038/s41541-023-00796-7] [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: 08/17/2023] [Accepted: 12/07/2023] [Indexed: 01/08/2024] Open
Abstract
The receptor-binding domain, region II, of the Plasmodium vivax Duffy binding protein (PvDBPII) binds the Duffy antigen on the reticulocyte surface to mediate invasion. A heterologous vaccine challenge trial recently showed that a delayed dosing regimen with recombinant PvDBPII SalI variant formulated with adjuvant Matrix-MTM reduced the in vivo parasite multiplication rate (PMR) in immunized volunteers challenged with the Thai P. vivax isolate PvW1. Here, we describe extensive analysis of the polyfunctional antibody responses elicited by PvDBPII immunization and identify immune correlates for PMR reduction. A classification algorithm identified antibody features that significantly contribute to PMR reduction. These included antibody titre, receptor-binding inhibitory titre, dissociation constant of the PvDBPII-antibody interaction, complement C1q and Fc gamma receptor binding and specific IgG subclasses. These data suggest that multiple immune mechanisms elicited by PvDBPII immunization are likely to be associated with protection and the immune correlates identified could guide the development of an effective vaccine for P. vivax malaria. Importantly, all the polyfunctional antibody features that correlated with protection cross-reacted with both PvDBPII SalI and PvW1 variants, suggesting that immunization with PvDBPII should protect against diverse P. vivax isolates.
Collapse
Affiliation(s)
- Francisco J Martinez
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, Paris, France
| | - Michael White
- Infectious Disease Epidemiology and Analytics G5 Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | | | - Christèle Huon
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, Paris, France
| | - Alix Boucharlat
- Chemogenomic and Biological Screening Core Facility, C2RT, Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Paris, France
| | - Fabrice Agou
- Chemogenomic and Biological Screening Core Facility, C2RT, Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Paris, France
| | - Patrick England
- Molecular Biophysics Facility, CNRS UMR 3528, Institut Pasteur, Paris, France
| | - Jean Popovici
- Malaria Reasearch Unit, Institut Pasteur du Cambodge, Pnom Penh, Cambodia
| | - Mimi M Hou
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
| | - Sarah E Silk
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
| | - Jordan R Barrett
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
| | - Carolyn M Nielsen
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
| | | | | | - Virander S Chauhan
- International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Angela M Minassian
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Chetan E Chitnis
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, Paris, France.
| |
Collapse
|
16
|
Kibwana E, Kapulu M. Controlled Human Malaria Infection Studies in Africa-Past, Present, and Future. Curr Top Microbiol Immunol 2024; 445:337-365. [PMID: 35704094 PMCID: PMC7616462 DOI: 10.1007/82_2022_256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Controlled human infection studies have contributed significantly to the understanding of pathogeneses and treatment of infectious diseases. In malaria, deliberately infecting humans with malaria parasites was used as a treatment for neurosyphilis in the early 1920s. More recently, controlled human malaria infection (CHMI) has become a valuable, cost-effective tool to fast-track the development and evaluation of new anti-malarial drugs and/or vaccines. CHMI studies have also been used to define host/parasite interactions and immunological correlates of protection. CHMI involves infecting a small number of healthy volunteers with malaria parasites, monitoring their parasitemia and providing anti-malarial treatment when a set threshold is reached. In this review we discuss the introduction, development, and challenges of modern-day Plasmodium falciparum CHMI studies conducted in Africa, and the impact of naturally acquired immunity on infectivity and vaccine efficacy. CHMIs have shown to be an invaluable tool particularly in accelerating malaria vaccine research. Although there are limitations of CHMI studies for estimating public health impacts and for regulatory purposes, their strength lies in proof-of-concept efficacy data at an early stage of development, providing a faster way to select vaccines for further development and providing valuable insights in understanding the mechanisms of immunity to malarial infection.
Collapse
Affiliation(s)
- Elizabeth Kibwana
- Bioscience Department, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | - Melissa Kapulu
- Bioscience Department, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| |
Collapse
|
17
|
Okoth WA, Ho MF, Zaman M, Cooper E, Som P, Burgess M, Walton M, Nevagi RJ, Beattie L, Murphy D, Stanisic DI, Good MF. A CAF01-adjuvanted whole asexual blood-stage liposomal malaria vaccine induces a CD4 + T-cell-dependent strain-transcending protective immunity in rodent models. mBio 2023; 14:e0254723. [PMID: 37962347 PMCID: PMC10746282 DOI: 10.1128/mbio.02547-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 11/15/2023] Open
Abstract
IMPORTANCE Malaria is a devastating disease that has claimed many lives, especially children <5 years of age in Sub-Saharan Africa, as documented in World Malaria Reports by WHO. Even though vector control and chemoprevention tools have helped with elimination efforts in some, if not all, endemic areas, these efforts have been hampered by serious issues (including drug and insecticide resistance and disruption to social cohesion caused by the COVID-19 pandemic). Development of an effective malaria vaccine is the alternative preventative tool in the fight against malaria. Vaccines save millions of lives each year and have helped in elimination and/or eradication of global diseases. Development of a highly efficacious malaria vaccine that will ensure long-lasting protective immunity will be a "game-changing" prevention strategy to finally eradicate the disease. Such a vaccine will need to counteract the significant obstacles that have been hampering subunit vaccine development to date, including antigenic polymorphism, sub-optimal immunogenicity, and waning vaccine efficacy.
Collapse
Affiliation(s)
- Winter A. Okoth
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Mei-Fong Ho
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Mehfuz Zaman
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Emily Cooper
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Priyanka Som
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Mark Burgess
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Maddison Walton
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Reshma J. Nevagi
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Lynette Beattie
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Declan Murphy
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Michael F. Good
- Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| |
Collapse
|
18
|
Abstract
Malaria is a mosquito-borne disease caused by protozoan parasites of the genus Plasmodium. Despite significant declines in malaria-attributable morbidity and mortality over the last two decades, it remains a major public health burden in many countries. This underscores the critical need for improved strategies to prevent, treat and control malaria if we are to ultimately progress towards the eradication of this disease. Ideally, this will include the development and deployment of a highly effective malaria vaccine that is able to induce long-lasting protective immunity. There are many malaria vaccine candidates in development, with more than a dozen of these in clinical development. RTS,S/AS01 (also known as Mosquirix) is the most advanced malaria vaccine and was shown to have modest efficacy against clinical malaria in phase III trials in 5- to 17-month-old infants. Following pilot implementation trials, the World Health Organisation has recommended it for use in Africa in young children who are most at risk of infection with P. falciparum, the deadliest of the human malaria parasites. It is well recognised that more effective malaria vaccines are needed. In this review, we discuss malaria vaccine candidates that have progressed into clinical evaluation and highlight the most advanced candidates: Sanaria's irradiated sporozoite vaccine (PfSPZ Vaccine), the chemoattenuated sporozoite vaccine (PfSPZ-CVac), RTS,S/AS01 and the novel malaria vaccine candidate, R21, which displayed promising, high-level efficacy in a recent small phase IIb trial in Africa.
Collapse
Affiliation(s)
- Danielle I Stanisic
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| | - Michael F Good
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| |
Collapse
|
19
|
Amai M, Nojima M, Yuki Y, Kiyono H, Nagamura F. A review of criteria strictness in "Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials". Vaccine 2023; 41:5622-5629. [PMID: 37532612 DOI: 10.1016/j.vaccine.2023.07.072] [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: 06/28/2022] [Revised: 07/25/2023] [Accepted: 07/29/2023] [Indexed: 08/04/2023]
Abstract
To assess safety in vaccine development, stricter grading scales, such as the "Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials" issued by the U.S. Food and Drug Administration (FDA grading scale), are required. However, concern exists that their strictness may lead to an overestimation of some adverse events (AEs). We analyzed the details of AEs in a phase I clinical trial of a preventive vaccine for infectious diseases. In this trial, we observed the high occurrence of Grade 1 or greater AEs in hemoglobin changes from baseline value, and hypernatremia, and hypokalemia by FDA grading scale. The range considered as non-AE according to the FDA grading scale shifted or became narrower when compared to reference intervals, especially for a Japanese cohort. For sodium grading, the criterion for hypernatremia was around 2 to mEq/L lower than the upper limit of most standards in several countries. Also, the criterion for hypokalemia was around 0.2 mEq/L higher than the lower limit of most standards. Regarding a decrease in hemoglobin from baseline, the criterion of "any decrease" used for a Grade 1 AE was too strict and we suggest this be omitted. Upper and lower limits of AE criteria for sodium and potassium should be equal to, or 10-20% above, the reference interval consistent with other toxicities determined by laboratory tests. Consideration should be given to the issues surrounding the criteria that determine AEs before conducting clinical trials.
Collapse
Affiliation(s)
- Motoki Amai
- Center for Translational Research, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masanori Nojima
- Center for Translational Research, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Advanced Medicine Promotion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Yoshikazu Yuki
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; HanaVax Inc., Chiba, Japan
| | - Hiroshi Kiyono
- HanaVax Inc., Chiba, Japan; Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Future Medicine Education and Research Organization, Chiba University, Chiba, Japan; CU-UCSD Center for Mucosal Immunology, Allergy, and Vaccine (cMAV), Departments of Medicine and Pathology, University of California, San Diego, CA, USA
| | - Fumitaka Nagamura
- Center for Translational Research, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Advanced Medicine Promotion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
20
|
Martinez FJ, Guillotte-Blisnick M, Huon C, England P, Popovici J, Laude H, Arowas L, Ungeheuer MN, Reimer JM, Carter D, Reed S, Mukherjee P, Chauhan VS, Chitnis CE. Immunogenicity of a Plasmodium vivax vaccine based on the duffy binding protein formulated using adjuvants compatible for use in humans. Sci Rep 2023; 13:13904. [PMID: 37626150 PMCID: PMC10457348 DOI: 10.1038/s41598-023-40043-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The invasion of reticulocytes by Plasmodium vivax merozoites is dependent on the interaction of the Plasmodium vivax Duffy Binding Protein (PvDBP) with the Duffy antigen receptor for chemokines (DARC). The N-terminal cysteine-rich region II of PvDBP (PvDBPII), which binds DARC, is a leading P. vivax malaria vaccine candidate. Here, we have evaluated the immunogenicity of recombinant PvDBPII formulated with the adjuvants Matrix-M and GLA-SE in mice. Analysis of the antibody responses revealed comparable ELISA recognition titres as well as similar recognition of native PvDBP in P. vivax schizonts by immunofluorescence assay. Moreover, antibodies elicited by the two adjuvant formulations had similar functional properties such as avidity, isotype profile and inhibition of PvDBPII-DARC binding. Furthermore, the anti-PvDBPII antibodies were able to block the interaction of DARC with the homologous PvDBPII SalI allele as well as the heterologous PvDBPII PvW1 allele from a Thai clinical isolate that is used for controlled human malaria infections (CHMI). The cross-reactivity of these antibodies with PvW1 suggest that immunization with the PvDBPII SalI strain should neutralize reticulocyte invasion by the challenge P. vivax strain PvW1.
Collapse
Affiliation(s)
- Francisco J Martinez
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, 25-28 Rue du Dr. Roux, 75015, Paris, France
| | - Micheline Guillotte-Blisnick
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, 25-28 Rue du Dr. Roux, 75015, Paris, France
| | - Christèle Huon
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, 25-28 Rue du Dr. Roux, 75015, Paris, France
| | - Patrick England
- Plate-Forme de Biophysique Moléculaire, CNRS UMR 3528, Institut Pasteur, Université Paris Cité, Paris, France
| | - Jean Popovici
- Malaria Research Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Hélène Laude
- Investigational Clinical Service and Access to Research Bio-Resources (ICAReB), Institut Pasteur, Paris, France
| | - Laurence Arowas
- Investigational Clinical Service and Access to Research Bio-Resources (ICAReB), Institut Pasteur, Paris, France
| | - Marie-Noëlle Ungeheuer
- Investigational Clinical Service and Access to Research Bio-Resources (ICAReB), Institut Pasteur, Paris, France
| | | | - Darrick Carter
- HDT Bio, Seattle, WA, USA
- PAI Life Sciences Inc., Seattle, WA, USA
| | | | | | - Virander S Chauhan
- International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Chetan E Chitnis
- Unité de Biologie de Plasmodium et Vaccins, Institut Pasteur, Université Paris Cité, 25-28 Rue du Dr. Roux, 75015, Paris, France.
| |
Collapse
|
21
|
Müllertz OAO, Andersen P, Christensen D, Foged C, Thakur A. Pulmonary Administration of the Liposome-Based Adjuvant CAF01: Effect of Surface Charge on Mucosal Adjuvant Function. Mol Pharm 2023; 20:953-970. [PMID: 36583936 DOI: 10.1021/acs.molpharmaceut.2c00634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mucosal surfaces of the lungs represent a major site of entry for airborne pathogens, and pulmonary administration of vaccines is an attractive strategy to induce protective mucosal immunity in the airways. Recently, we demonstrated the potential of pulmonary vaccination with the tuberculosis subunit antigen H56 adjuvanted with the cationic liposomal adjuvant formulation CAF01, which consists of the cationic lipid dimethyldioctadecylammonium (DDA) bromide and the synthetic cord factor trehalose-6,6'-dibehenate. However, the cationic charge of DDA represents a major safety challenge. Hence, replacing DDA with a safer zwitterionic or anionic phospholipid is an attractive approach to improve vaccine safety, but the effect of liposomal surface charge on the induction of mucosal immunity after airway immunization is poorly understood. Here, we investigated the effect of surface charge by replacing the cationic DDA component of CAF01 with zwitterionic dipalmitoylphosphatidylcholine (DPPC) or anionic dipalmitoylphosphatidylglycerol (DPPG), and we show that charge modification enhances antigen-specific pulmonary T-cell responses against co-formulated H56. We systematically replaced DDA with either DPPC or DPPG and found that these modifications resulted in colloidally stable liposomes that have similar size and morphology to unmodified CAF01. DPPC- or DPPG-modified CAF01 displayed surface charge-dependent protein adsorption and induced slightly higher follicular helper T cells and germinal center B cells in the lung-draining lymph nodes than unmodified CAF01. In addition, modified CAF01 induced significantly higher levels of H56-specific Th17 cells and polyfunctional CD4+ T cells in the lungs, as compared to unmodified CAF01. However, the strong H56-specific humoral responses induced by CAF01 in the lungs and spleen were not influenced by surface charge. Hence, these results provide insights into the importance of surface charge for liposomal adjuvant function and can also guide the design of safe pulmonary subunit vaccines against other mucosal pathogens.
Collapse
Affiliation(s)
- Olivia Amanda Oest Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen Ø2100, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, Copenhagen S2300, Denmark
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, Copenhagen S2300, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen Ø2100, Denmark
| | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, Copenhagen Ø2100, Denmark
| |
Collapse
|
22
|
Teymournejad O, Li Z, Beesetty P, Yang C, Montgomery CP. Toxin expression during Staphylococcus aureus infection imprints host immunity to inhibit vaccine efficacy. NPJ Vaccines 2023; 8:3. [PMID: 36693884 PMCID: PMC9873725 DOI: 10.1038/s41541-022-00598-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/05/2022] [Indexed: 01/26/2023] Open
Abstract
Staphylococcus aureus infections are a major public health issue, and a vaccine is urgently needed. Despite a considerable promise in preclinical models, all vaccines tested thus far have failed to protect humans against S. aureus. Unlike laboratory mice, humans are exposed to S. aureus throughout life. In the current study, we hypothesized that prior exposure to S. aureus "imprints" the immune response to inhibit vaccine-mediated protection. We established a mouse model in which S. aureus skin and soft tissue infection (SSTI) is followed by vaccination and secondary SSTI. Unlike naïve mice, S. aureus-sensitized mice were incompletely protected against secondary SSTI by vaccination with the inactivated α-hemolysin (Hla) mutant HlaH35L. Inhibition of protection was specific for the HlaH35L vaccine and required hla expression during primary SSTI. Surprisingly, inhibition occurred at the level of vaccine-elicited effector T cells; hla expression during primary infection limited the expansion of T cells and dendritic cells and impaired vaccine-specific T cell responses. Importantly, the T cell-stimulating adjuvant CAF01 rescued inhibition and restored vaccine-mediated protection. Together, these findings identify a potential mechanism for the failure of translation of promising S. aureus vaccines from mouse models to clinical practice and suggest a path forward to prevent these devastating infections.
Collapse
Affiliation(s)
- Omid Teymournejad
- grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH US ,grid.185648.60000 0001 2175 0319Present Address: Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL US
| | - Zhaotao Li
- grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH US
| | - Pavani Beesetty
- grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH US ,grid.231844.80000 0004 0474 0428Present Address: Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario Canada
| | - Ching Yang
- grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH US ,grid.259180.70000 0001 2298 1899Present Address: Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY US
| | - Christopher P. Montgomery
- grid.240344.50000 0004 0392 3476Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH US ,grid.261331.40000 0001 2285 7943Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH US ,grid.240344.50000 0004 0392 3476Division of Critical Care Medicine, Nationwide Children’s Hospital, Columbus, OH US
| |
Collapse
|
23
|
Chandley P, Ranjan R, Kumar S, Rohatgi S. Host-parasite interactions during Plasmodium infection: Implications for immunotherapies. Front Immunol 2023; 13:1091961. [PMID: 36685595 PMCID: PMC9845897 DOI: 10.3389/fimmu.2022.1091961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Malaria is a global infectious disease that remains a leading cause of morbidity and mortality in the developing world. Multiple environmental and host and parasite factors govern the clinical outcomes of malaria. The host immune response against the Plasmodium parasite is heterogenous and stage-specific both in the human host and mosquito vector. The Plasmodium parasite virulence is predominantly associated with its ability to evade the host's immune response. Despite the availability of drug-based therapies, Plasmodium parasites can acquire drug resistance due to high antigenic variations and allelic polymorphisms. The lack of licensed vaccines against Plasmodium infection necessitates the development of effective, safe and successful therapeutics. To design an effective vaccine, it is important to study the immune evasion strategies and stage-specific Plasmodium proteins, which are targets of the host immune response. This review provides an overview of the host immune defense mechanisms and parasite immune evasion strategies during Plasmodium infection. Furthermore, we also summarize and discuss the current progress in various anti-malarial vaccine approaches, along with antibody-based therapy involving monoclonal antibodies, and research advancements in host-directed therapy, which can together open new avenues for developing novel immunotherapies against malaria infection and transmission.
Collapse
Affiliation(s)
- Pankaj Chandley
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Ravikant Ranjan
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Soma Rohatgi
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India,*Correspondence: Soma Rohatgi,
| |
Collapse
|
24
|
Singh SK, Naghizadeh M, Plieskatt J, Singh S, Theisen M. Cloning and Recombinant Protein Expression in Lactococcus lactis. Methods Mol Biol 2023; 2652:3-20. [PMID: 37093467 DOI: 10.1007/978-1-0716-3147-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The Lactococcus lactis, a Gram-positive bacteria, is an ideal expression host for the overproduction of heterologous proteins in a properly folded and functional form. L. lactis has been identified as an efficient cell factory, generally recognized as safe (GRAS), has a long history of safe use in food production, and is known to have probiotic properties. Key desirable features of L. lactis include the following: (1) rapid growth to high cell densities, not requiring aeration which facilitates large-scale fermentation; (2) its Gram-positive nature precludes the presence of contaminating endotoxins; (3) the capacity to secrete stable recombinant protein into the growth medium with few proteases resulting in a properly folded, full-length protein; and (4) the availability of diverse expression vectors facilitating various cloning options. We have previously described production of several recombinant proteins with varying degrees of predicted structural complexities using the L. lactis pH-dependent P170 promoter. The purpose of this chapter is to provide a detailed protocol for facilitating wider application of L. lactis as a reliable platform for expression of heterologous recombinant proteins in soluble form. Here, we present details of the various steps involved such as cloning of the target gene in appropriate expression plasmid vector, determination of the expression levels of the heterologous protein, and initial purification of the expressed soluble recombinant protein of interest.
Collapse
Affiliation(s)
- Susheel K Singh
- Biotherapeutic and Vaccine Research Division, ICMR-Regional Medical Research Centre, Bhubaneswar, Odisha, India
| | - Mohammad Naghizadeh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jordan Plieskatt
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Subhash Singh
- Biotherapeutic and Vaccine Research Division, ICMR-Regional Medical Research Centre, Bhubaneswar, Odisha, India
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Richie TL, Church LWP, Murshedkar T, Billingsley PF, James ER, Chen MC, Abebe Y, KC N, Chakravarty S, Dolberg D, Healy SA, Diawara H, Sissoko MS, Sagara I, Cook DM, Epstein JE, Mordmüller B, Kapulu M, Kreidenweiss A, Franke-Fayard B, Agnandji ST, López Mikue MSA, McCall MBB, Steinhardt L, Oneko M, Olotu A, Vaughan AM, Kublin JG, Murphy SC, Jongo S, Tanner M, Sirima SB, Laurens MB, Daubenberger C, Silva JC, Lyke KE, Janse CJ, Roestenberg M, Sauerwein RW, Abdulla S, Dicko A, Kappe SHI, Lee Sim BK, Duffy PE, Kremsner PG, Hoffman SL. Sporozoite immunization: innovative translational science to support the fight against malaria. Expert Rev Vaccines 2023; 22:964-1007. [PMID: 37571809 PMCID: PMC10949369 DOI: 10.1080/14760584.2023.2245890] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
INTRODUCTION Malaria, a devastating febrile illness caused by protozoan parasites, sickened 247,000,000 people in 2021 and killed 619,000, mostly children and pregnant women in sub-Saharan Africa. A highly effective vaccine is urgently needed, especially for Plasmodium falciparum (Pf), the deadliest human malaria parasite. AREAS COVERED Sporozoites (SPZ), the parasite stage transmitted by Anopheles mosquitoes to humans, are the only vaccine immunogen achieving >90% efficacy against Pf infection. This review describes >30 clinical trials of PfSPZ vaccines in the U.S.A., Europe, Africa, and Asia, based on first-hand knowledge of the trials and PubMed searches of 'sporozoites,' 'malaria,' and 'vaccines.' EXPERT OPINION First generation (radiation-attenuated) PfSPZ vaccines are safe, well tolerated, 80-100% efficacious against homologous controlled human malaria infection (CHMI) and provide 18-19 months protection without boosting in Africa. Second generation chemo-attenuated PfSPZ are more potent, 100% efficacious against stringent heterologous (variant strain) CHMI, but require a co-administered drug, raising safety concerns. Third generation, late liver stage-arresting, replication competent (LARC), genetically-attenuated PfSPZ are expected to be both safe and highly efficacious. Overall, PfSPZ vaccines meet safety, tolerability, and efficacy requirements for protecting pregnant women and travelers exposed to Pf in Africa, with licensure for these populations possible within 5 years. Protecting children and mass vaccination programs to block transmission and eliminate malaria are long-term objectives.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sara A. Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Halimatou Diawara
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Mahamadou S. Sissoko
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Issaka Sagara
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - David M. Cook
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Judith E. Epstein
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Mordmüller
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Melissa Kapulu
- Biosciences Department, Kenya Medical Research Institute KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrea Kreidenweiss
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | | | - Selidji T. Agnandji
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | | - Matthew B. B. McCall
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Laura Steinhardt
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Martina Oneko
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Ally Olotu
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Ashley M. Vaughan
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - James G. Kublin
- Department of Global Health, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases and Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Said Jongo
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Marcel Tanner
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Claudia Daubenberger
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Joana C. Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chris J. Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert W. Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Salim Abdulla
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Alassane Dicko
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - 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
| | | | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter G. Kremsner
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | |
Collapse
|
26
|
Takashima E, Nagaoka H, Correia R, Alves PM, Roldão A, Christensen D, Guderian JA, Fukushima A, Viebig NK, Depraetere H, Tsuboi T. A novel asexual blood-stage malaria vaccine candidate: PfRipr5 formulated with human-use adjuvants induces potent growth inhibitory antibodies. Front Immunol 2022; 13:1002430. [PMID: 36389677 PMCID: PMC9647036 DOI: 10.3389/fimmu.2022.1002430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2023] Open
Abstract
PfRipr is a highly conserved asexual-blood stage malaria vaccine candidate against Plasmodium falciparum. PfRipr5, a protein fragment of PfRipr inducing the most potent inhibitory antibodies, is a promising candidate for the development of next-generation malaria vaccines, requiring validation of its potential when formulated with adjuvants already approved for human use. In this study, PfRipr5 antigen was efficiently produced in a tank bioreactor using insect High Five cells and the baculovirus expression vector system; purified PfRipr5 was thermally stable in its monomeric form, had high purity and binding capacity to functional monoclonal anti-PfRipr antibody. The formulation of purified PfRipr5 with Alhydrogel®, GLA-SE or CAF®01 adjuvants accepted for human use showed acceptable compatibility. Rabbits immunized with these formulations induced comparable levels of anti-PfRipr5 antibodies, and significantly higher than the control group immunized with PfRipr5 alone. To investigate the efficacy of the antibodies, we used an in vitro parasite growth inhibition assay (GIA). The highest average GIA activity amongst all groups was attained with antibodies induced by immunization with PfRipr5 formulated with CAF®01. Overall, this study validates the potential of adjuvanted PfRipr5 as an asexual blood-stage malaria vaccine candidate, with PfRipr5/CAF®01 being a promising formulation for subsequent pre-clinical and clinical development.
Collapse
Affiliation(s)
- Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Ricardo Correia
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - António Roldão
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut (SSI), Copenhagen, Denmark
| | | | | | - Nicola K. Viebig
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Hilde Depraetere
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
27
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
28
|
Owalla TJ, Hergott DEB, Seilie AM, Staubus W, Chavtur C, Chang M, Kublin JG, Egwang TG, Murphy SC. Rethinking detection of pre-existing and intervening Plasmodium infections in malaria clinical trials. Front Immunol 2022; 13:1003452. [PMID: 36203582 PMCID: PMC9531235 DOI: 10.3389/fimmu.2022.1003452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/22/2022] [Indexed: 02/02/2023] Open
Abstract
Pre-existing and intervening low-density Plasmodium infections complicate the conduct of malaria clinical trials. These infections confound infection detection endpoints, and their immunological effects may detract from intended vaccine-induced immune responses. Historically, these infections were often unrecognized since infrequent and often analytically insensitive parasitological testing was performed before and during trials. Molecular diagnostics now permits their detection, but investigators must weigh the cost, complexity, and personnel demands on the study and the laboratory when scheduling such tests. This paper discusses the effect of pre-existing and intervening, low-density Plasmodium infections on malaria vaccine trial endpoints and the current methods employed for their infection detection. We review detection techniques, that until recently, provided a dearth of cost-effective strategies for detecting low density infections. A recently deployed, field-tested, simple, and cost-effective molecular diagnostic strategy for detecting pre-existing and intervening Plasmodium infections from dried blood spots (DBS) in malaria-endemic settings is discussed to inform new clinical trial designs. Strategies that combine sensitive molecular diagnostic techniques with convenient DBS collections and cost-effective pooling strategies may enable more thorough and informative infection monitoring in upcoming malaria clinical trials and epidemiological studies.
Collapse
Affiliation(s)
- Tonny J. Owalla
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Dianna E. B. Hergott
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
| | - Annette M. Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Weston Staubus
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Chris Chavtur
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Ming Chang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - James G. Kublin
- Department of Global Health, University of Washington, Seattle, WA, United States,Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Thomas G. Egwang
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States,Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States,Department of Microbiology, University of Washington, Seattle, WA, United States,*Correspondence: Sean C. Murphy,
| |
Collapse
|
29
|
Chakravarty S, Shears MJ, James ER, Rai U, Kc N, Conteh S, Lambert LE, Duffy PE, Murphy SC, Hoffman SL. Efficient infection of non-human primates with purified, cryopreserved Plasmodium knowlesi sporozoites. Malar J 2022; 21:247. [PMID: 36030292 PMCID: PMC9418655 DOI: 10.1186/s12936-022-04261-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/17/2022] [Indexed: 11/29/2022] Open
Abstract
Background Plasmodium falciparum (Pf) sporozoite (SPZ) vaccines are the only candidate malaria vaccines that induce > 90% vaccine efficacy (VE) against controlled human malaria infection and the only malaria vaccines to have achieved reproducible VE against malaria in adults in Africa. The goal is to increase the impact and reduce the cost of PfSPZ vaccines by optimizing vaccine potency and manufacturing, which will benefit from identification of immunological responses contributing to protection in humans. Currently, there is no authentic animal challenge model for assessing P. falciparum malaria VE. Alternatively, Plasmodium knowlesi (Pk), which infects humans and non-human primates (NHPs) in nature, can be used to experimentally infect rhesus macaques (Macaca mulatta) to assess VE. Methods Sanaria has, therefore, produced purified, vialed, cryopreserved PkSPZ and conducted challenge studies in several naïve NHP cohorts. In the first cohort, groups of three rhesus macaques each received doses of 5 × 102, 2.5 × 103, 1.25 × 104 and 2.5 × 104 PkSPZ administered by direct venous inoculation. The infectivity of 1.5 × 103 PkSPZ cryopreserved with an altered method and of 1.5 × 103 PkSPZ cryopreserved for four years was tested in a second and third cohort of rhesus NHPs. The lastly, three pig-tailed macaques (Macaca nemestrina), a natural P. knowlesi host, were challenged with 2.5 × 103 PkSPZ cryopreserved six years earlier. Results In the first cohort, all 12 animals developed P. knowlesi parasitaemia by thick blood smear, and the time to positivity (prepatent period) followed a non-linear 4-parameter logistic sigmoidal model with a median of 11, 10, 8, and 7 days, respectively (r2 = 1). PkSPZ cryopreserved using a modified rapid-scalable method infected rhesus with a pre-patent period of 10 days, as did PkSPZ cryopreserved four years prior to infection, similar to the control group. Cryopreserved PkSPZ infected pig-tailed macaques with median time to positivity by thin smear, of 11 days. Conclusion This study establishes the capacity to consistently infect NHPs with purified, vialed, cryopreserved PkSPZ, providing a foundation for future studies to probe protective immunological mechanisms elicited by PfSPZ vaccines that cannot be established in humans. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04261-z.
Collapse
Affiliation(s)
- Sumana Chakravarty
- Sanaria, Inc, 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Melanie J Shears
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.,Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | - Eric R James
- Sanaria, Inc, 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Urvashi Rai
- Sanaria, Inc, 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Natasha Kc
- Sanaria, Inc, 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA
| | - Solomon Conteh
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, USA
| | - Lynn E Lambert
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, Bethesda, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.,Washington National Primate Research Center, University of Washington, Seattle, WA, USA.,Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Stephen L Hoffman
- Sanaria, Inc, 9800 Medical Center Drive, Suite A209, Rockville, MD, 20850, USA.
| |
Collapse
|
30
|
Takashima E, Kanoi BN, Nagaoka H, Morita M, Hassan I, Palacpac NMQ, Egwang TG, Horii T, Gitaka J, Tsuboi T. Meta-Analysis of Human Antibodies Against Plasmodium falciparum Variable Surface and Merozoite Stage Antigens. Front Immunol 2022; 13:887219. [PMID: 35757771 PMCID: PMC9218060 DOI: 10.3389/fimmu.2022.887219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Concerted efforts to fight malaria have caused significant reductions in global malaria cases and mortality. Sustaining this will be critical to avoid rebound and outbreaks of seasonal malaria. Identifying predictive attributes that define clinical malaria will be key to guide development of second-generation tools to fight malaria. Broadly reactive antibodies against variable surface antigens that are expressed on the surface of infected erythrocytes and merozoites stage antigens are targets of naturally acquired immunity and prime candidates for anti-malaria therapeutics and vaccines. However, predicting the relationship between the antigen-specific antibodies and protection from clinical malaria remains unresolved. Here, we used new datasets and multiple approaches combined with re-analysis of our previous data to assess the multi-dimensional and complex relationship between antibody responses and clinical malaria outcomes. We observed 22 antigens (17 PfEMP1 domains, 3 RIFIN family members, merozoite surface protein 3 (PF3D7_1035400), and merozoites-associated armadillo repeats protein (PF3D7_1035900) that were selected across three different clinical malaria definitions (1,000/2,500/5,000 parasites/µl plus fever). In addition, Principal Components Analysis (PCA) indicated that the first three components (Dim1, Dim2 and Dim3 with eigenvalues of 306, 48, and 29, respectively) accounted for 66.1% of the total variations seen. Specifically, the Dim1, Dim2 and Dim3 explained 52.8%, 8.2% and 5% of variability, respectively. We further observed a significant relationship between the first component scores and age with antibodies to PfEMP1 domains being the key contributing variables. This is consistent with a recent proposal suggesting that there is an ordered acquisition of antibodies targeting PfEMP1 proteins. Thus, although limited, and further work on the significance of the selected antigens will be required, these approaches may provide insights for identification of drivers of naturally acquired protective immunity as well as guide development of additional tools for malaria elimination and eradication.
Collapse
Affiliation(s)
- Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Bernard N Kanoi
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Ifra Hassan
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Nirianne M Q Palacpac
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | | | - Toshihiro Horii
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Jesse Gitaka
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
31
|
Nouatin O, Ibáñez J, Fendel R, Ngoa UA, Lorenz FR, Dejon-Agobé JC, Edoa JR, Flügge J, Brückner S, Esen M, Theisen M, Hoffman SL, Moutairou K, Luty AJF, Lell B, Kremsner PG, Adegnika AA, Mordmüller B. Cellular and antibody response in GMZ2-vaccinated Gabonese volunteers in a controlled human malaria infection trial. Malar J 2022; 21:191. [PMID: 35715803 PMCID: PMC9204906 DOI: 10.1186/s12936-022-04169-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Antibody and cellular memory responses following vaccination are important measures of immunogenicity. These immune markers were quantified in the framework of a vaccine trial investigating the malaria vaccine candidate GMZ2. METHODS Fifty Gabonese adults were vaccinated with two formulations (aluminum Alhydrogel and CAF01) of GMZ2 or a control vaccine (Verorab). Vaccine efficacy was assessed using controlled human malaria infection (CHMI) by direct venous inoculation of 3200 live Plasmodium falciparum sporozoites (PfSPZ Challenge). GMZ2-stimulated T and specific B-cell responses were estimated by flow cytometry before and after vaccination. Additionally, the antibody response against 212 P. falciparum antigens was estimated before CHMI by protein microarray. RESULTS Frequencies of pro- and anti-inflammatory CD4+ T cells stimulated with the vaccine antigen GMZ2 as well as B cell profiles did not change after vaccination. IL-10-producing CD4+ T cells and CD20+ IgG+ B cells were increased post-vaccination regardless of the intervention, thus could not be specifically attributed to any malaria vaccine regimen. In contrast, GMZ2-specific antibody response increased after the vaccination, but was not correlated to protection. Antibody responses to several P. falciparum blood and liver stage antigens (MSP1, MSP4, MSP8, PfEMP1, STARP) as well as the breadth of the malaria-specific antibody response were significantly higher in protected study participants. CONCLUSIONS In lifelong malaria exposed adults, the main marker of protection against CHMI is a broad antibody pattern recognizing multiple stages of the plasmodial life cycle. Despite vaccination with GMZ2 using a novel formulation, expansion of the GMZ2-stimulated T cells or the GMZ2-specific B cell response was limited, and the vaccine response could not be identified as a marker of protection against malaria. Trial registration PACTR; PACTR201503001038304; Registered 17 February 2015; https://pactr.samrc.ac.za/TrialDisplay.aspx?TrialID=1038.
Collapse
Affiliation(s)
- Odilon Nouatin
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon. .,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany. .,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany. .,Département de Biochimie Et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d'Abomey-Calavi, Cotonou, Bénin.
| | - Javier Ibáñez
- Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany
| | - Rolf Fendel
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon. .,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany. .,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany.
| | - Ulysse A Ngoa
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon
| | - Freia-Raphaella Lorenz
- Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany
| | - Jean-Claude Dejon-Agobé
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jean Ronald Edoa
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany
| | - Judith Flügge
- Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany
| | - Sina Brückner
- Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany
| | - Meral Esen
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany.,Cluster of Excellence: EXC 2124: Controlling Microbes to Fight Infection, Tübingen, Germany
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | | | - Kabirou Moutairou
- Département de Biochimie Et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d'Abomey-Calavi, Cotonou, Bénin
| | - Adrian J F Luty
- Centre d'Etude et de Recherche sur le Paludisme Associé à la Grossesse et a l'Enfance, Calavi, Bénin.,MERIT, Université de Paris, Paris, IRD, France
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany
| | - Ayola A Adegnika
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany.,Department of Parasitology, Leiden University Medical Centre (LUMC), 2333 ZA, Leiden, The Netherlands.,Fondation pour la Recherche Scientifique, 72 BP45, Cotonou, Bénin
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany.,Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
32
|
Abstract
"The Primate Malarias" book has been a uniquely important resource for multiple generations of scientists, since its debut in 1971, and remains pertinent to the present day. Indeed, nonhuman primates (NHPs) have been instrumental for major breakthroughs in basic and pre-clinical research on malaria for over 50 years. Research involving NHPs have provided critical insights and data that have been essential for malaria research on many parasite species, drugs, vaccines, pathogenesis, and transmission, leading to improved clinical care and advancing research goals for malaria control, elimination, and eradication. Whilst most malaria scientists over the decades have been studying Plasmodium falciparum, with NHP infections, in clinical studies with humans, or using in vitro culture or rodent model systems, others have been dedicated to advancing research on Plasmodium vivax, as well as on phylogenetically related simian species, including Plasmodium cynomolgi, Plasmodium coatneyi, and Plasmodium knowlesi. In-depth study of these four phylogenetically related species over the years has spawned the design of NHP longitudinal infection strategies for gathering information about ongoing infections, which can be related to human infections. These Plasmodium-NHP infection model systems are reviewed here, with emphasis on modern systems biological approaches to studying longitudinal infections, pathogenesis, immunity, and vaccines. Recent discoveries capitalizing on NHP longitudinal infections include an advanced understanding of chronic infections, relapses, anaemia, and immune memory. With quickly emerging new technological advances, more in-depth research and mechanistic discoveries can be anticipated on these and additional critical topics, including hypnozoite biology, antigenic variation, gametocyte transmission, bone marrow dysfunction, and loss of uninfected RBCs. New strategies and insights published by the Malaria Host-Pathogen Interaction Center (MaHPIC) are recapped here along with a vision that stresses the importance of educating future experts well trained in utilizing NHP infection model systems for the pursuit of innovative, effective interventions against malaria.
Collapse
Affiliation(s)
- Mary R Galinski
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Vaccine Center, Emory University, Atlanta, GA, USA.
- Emory National Primate Research Center (Yerkes National Primate Research Center), Emory University, Atlanta, GA, USA.
| |
Collapse
|
33
|
Garcia‐del Rio L, Diaz‐Rodriguez P, Pedersen GK, Christensen D, Landin M. Sublingual Boosting with a Novel Mucoadhesive Thermogelling Hydrogel Following Parenteral CAF01 Priming as a Strategy Against Chlamydia trachomatis. Adv Healthc Mater 2022; 11:e2102508. [PMID: 35124896 PMCID: PMC11468966 DOI: 10.1002/adhm.202102508] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/18/2022] [Indexed: 01/13/2023]
Abstract
Chlamydia trachomatis is the most prevalent sexually transmitted disease of bacterial origin. The high number of asymptomatic cases makes it difficult to stop the transmission, requiring vaccine development. Herein, a strategy is proposed to obtain local genital tract immunity against C. trachomatis through parenteral prime and sublingual boost. Subcutaneous administration of chlamydia CTH522 subunit vaccine loaded in the adjuvant CAF01 is combined with sublingual administration of CTH522 loaded in a novel thermosensitive and mucoadhesive hydrogel. Briefly, a ternary optimized hydrogel (OGEL) with desirable biological and physicochemical properties is obtained using artificial intelligence techniques. This formulation exhibits a high gel strength and a strong mucoadhesive, adhesive and cohesive nature. The thermosensitive properties of the hydrogel facilitate application under the tongue. Meanwhile the fast gelation at body temperature together with rapid antigen release should avoid CTH522 leakage by swallowing and increase the contact with sublingual tissue, thus promoting absorption. In vivo studies demonstrate that parenteral-sublingual prime-boost immunization, using CAF01 and OGEL as CTH522 vaccine carriers, shows a tendency to increase cellular (Th1/Th17) immune responses when compared to mucosal or parenteral vaccination alone. Furthermore, parenteral prime with CAF01/CTH522 followed by sublingual boosting with OGEL/CTH522 elicits a local IgA response in the genital tract.
Collapse
Affiliation(s)
- Lorena Garcia‐del Rio
- Departamento de FarmacologíaFarmacia y Tecnología FarmacéuticaGrupo I+D Farma (GI‐1645)Agrupación Estratégica de Materiales (AeMat)Facultad de FarmaciaUniversidade de Santiago de CompostelaSantiago de Compostela15782Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)IDIS Research InstituteSantiago de Compostela15706Spain
| | - Patricia Diaz‐Rodriguez
- Departamento de FarmacologíaFarmacia y Tecnología FarmacéuticaGrupo I+D Farma (GI‐1645)Agrupación Estratégica de Materiales (AeMat)Facultad de FarmaciaUniversidade de Santiago de CompostelaSantiago de Compostela15782Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)IDIS Research InstituteSantiago de Compostela15706Spain
| | - Gabriel Kristian Pedersen
- Department of Infectious Disease ImmunologyStatens Serum InstitutArtillerivej 5Copenhagen S2300Denmark
| | - Dennis Christensen
- Department of Infectious Disease ImmunologyStatens Serum InstitutArtillerivej 5Copenhagen S2300Denmark
| | - Mariana Landin
- Departamento de FarmacologíaFarmacia y Tecnología FarmacéuticaGrupo I+D Farma (GI‐1645)Agrupación Estratégica de Materiales (AeMat)Facultad de FarmaciaUniversidade de Santiago de CompostelaSantiago de Compostela15782Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)IDIS Research InstituteSantiago de Compostela15706Spain
| |
Collapse
|
34
|
Dejon-Agobé JC, Edoa JR, Adegnika AA, Grobusch MP. Schistosomiasis in Gabon from 2000 to 2021 - A review. Acta Trop 2022; 228:106317. [PMID: 35051384 DOI: 10.1016/j.actatropica.2022.106317] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/20/2021] [Accepted: 01/16/2022] [Indexed: 11/01/2022]
Abstract
INTRODUCTION Schistosomiasis is a public health issue of concern in Gabon, with the disease being reported from all regions of the country. The topic has been of interest for the local researchers and physicians for over two decades. The objective of this narrative review was to provide an overview of the research activities in the area from 2000 to early 2021. METHODS We performed a narrative literature review. The search strategy was designed to get a broad overview of the different research topics on schistosomiasis and the national control programme, and included grey literature. RESULTS A total of 159 articles was screened, and 42 were included into the review in addition to the grey literature. During the past two decades, the work on schistosomiasis originated from five out of the nine provinces of the country, with diverse aspects of the disease investigated; including immunology, epidemiology, diagnosis and treatment. Several studies investigated various aspects of schistosomiasis-related morbidity in the respective study populations. The body of work demonstrates that much effort was made to understand the details of the host immune response to schistosomiasis, and the immune profile changes induced in patients treated with praziquantel. Although some MDA campaigns were conducted in the country; little, however, is known on the epidemiological situation of the disease, particularly of its distribution within the population, as well as co-infections with other parasitic diseases also endemic in the area. CONCLUSION Progress has been made over the past two decades in the understanding of schistosomiasis in the country, including disease-related morbidity and its interaction with other parasitic infections, and the immunology and epidemiology of the disease. However, for optimising control of the disease, there is a need to fine-tune these findings with detailed local epidemiological and malacological data. We call for such studies to accomplish the knowledge of schistosomiasis in the country, particularly in areas of moderate or high endemicity, and recommend this approach to comparable schistosomiasis-endemic areas elsewhere.
Collapse
|
35
|
Alves KCS, Guimarães JM, Almeida MEMD, Mariúba LAM. Plasmodium falciparum merozoite surface protein 3 as a vaccine candidate: a brief review. Rev Inst Med Trop Sao Paulo 2022; 64:e23. [PMID: 35293561 PMCID: PMC8916589 DOI: 10.1590/s1678-9946202264023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/24/2022] [Indexed: 11/22/2022] Open
Abstract
Despite the many efforts of researchers around the world, there is currently no effective vaccine for malaria. Numerous studies have been developed to find vaccine antigens that are immunogenic and safe. Among antigen candidates, Plasmodium falciparum merozoite surface protein 3 (MSP3) has stood out in a number of these studies for its ability to induce a consistent and protective immune response, also being safe for use in humans. This review presents the main studies that explored MSP3 as a vaccine candidate over the last few decades. MSP3 formulations were tested in animals and humans and the most advanced candidate formulations are MSP3-LSP, a combination of MSP3 and LSP1, and GMZ2 (a vaccine based on the recombinant protein fusion GLURP and MSP3) which is currently being tested in phase II clinical studies. This brief review highlights the history and the main formulations of MSP3-based vaccines approaches against P. falciparum .
Collapse
Affiliation(s)
| | | | | | - Luís André Morais Mariúba
- Instituto Leônidas e Maria Deane, Brazil; Universidade Federal do Amazonas, Brazil; Instituto Oswaldo Cruz, Brazil; Universidade Federal do Amazonas, Brazil
| |
Collapse
|
36
|
Immunosuppression in Malaria: Do Plasmodium falciparum Parasites Hijack the Host? Pathogens 2021; 10:pathogens10101277. [PMID: 34684226 PMCID: PMC8536967 DOI: 10.3390/pathogens10101277] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Malaria reflects not only a state of immune activation, but also a state of general immune defect or immunosuppression, of complex etiology that can last longer than the actual episode. Inhabitants of malaria-endemic regions with lifelong exposure to the parasite show an exhausted or immune regulatory profile compared to non- or minimally exposed subjects. Several studies and experiments to identify and characterize the cause of this malaria-related immunosuppression have shown that malaria suppresses humoral and cellular responses to both homologous (Plasmodium) and heterologous antigens (e.g., vaccines). However, neither the underlying mechanisms nor the relative involvement of different types of immune cells in immunosuppression during malaria is well understood. Moreover, the implication of the parasite during the different stages of the modulation of immunity has not been addressed in detail. There is growing evidence of a role of immune regulators and cellular components in malaria that may lead to immunosuppression that needs further research. In this review, we summarize the current evidence on how malaria parasites may directly and indirectly induce immunosuppression and investigate the potential role of specific cell types, effector molecules and other immunoregulatory factors.
Collapse
|
37
|
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
|
38
|
Nouatin O, Mengue JB, Dejon-Agobé JC, Fendel R, Ibáñez J, Ngoa UA, Edoa JR, Adégbité BR, Honkpéhédji YJ, Zinsou JF, Hounkpatin AB, Moutairou K, Homoet A, Esen M, Kreidenweiss A, Hoffman SL, Theisen M, Luty AJF, Lell B, Agnandji ST, Mombo-Ngoma G, Ramharter M, Kremsner P, Mordmüller B, Adegnika AA. Exploratory analysis of the effect of helminth infection on the immunogenicity and efficacy of the asexual blood-stage malaria vaccine candidate GMZ2. PLoS Negl Trop Dis 2021; 15:e0009361. [PMID: 34061838 PMCID: PMC8195366 DOI: 10.1371/journal.pntd.0009361] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 06/11/2021] [Accepted: 04/03/2021] [Indexed: 11/18/2022] Open
Abstract
Background Helminths can modulate the host immune response to Plasmodium falciparum and can therefore affect the risk of clinical malaria. We assessed here the effect of helminth infections on both the immunogenicity and efficacy of the GMZ2 malaria vaccine candidate, a recombinant protein consisting of conserved domains of GLURP and MSP3, two asexual blood-stage antigens of P. falciparum. Controlled human malaria infection (CHMI) was used to assess the efficacy of the vaccine. Methodology In a randomized, double-blind Phase I clinical trial, fifty, healthy, lifelong malaria-exposed adult volunteers received three doses of GMZ2 adjuvanted with either Cationic Adjuvant Formulation (CAF) 01 or Alhydrogel, or a control vaccine (Rabies) on days (D) 0, D28 and D56, followed by direct venous inoculation (DVI) of 3,200 P. falciparum sporozoites (PfSPZ Challenge) approximately 13 weeks after last vaccination to assess vaccine efficacy. Participants were followed-up on a daily basis with clinical examinations and thick blood smears to monitor P. falciparum parasitemia for 35 days. Malaria was defined as the presence of P. falciparum parasites in the blood associated with at least one symptom that can be associated to malaria over 35 days following DVI of PfSPZ Challenge. Soil-transmitted helminth (STH) infection was assessed by microscopy and by polymerase chain reaction (PCR) on stool, and Schistosoma infection was assessed by microscopy on urine. Participants were considered as infected if positive for any helminth either by PCR and/or microscopy at D0 and/or at D84 (Helm+) and were classified as mono-infection or co-infection. Total vaccine-specific IgG concentrations assessed on D84 were analysed as immunogenicity outcome. Main findings The helminth in mono-infection, particularly Schistosoma haematobium and STH were significantly associated with earlier malaria episodes following CHMI, while no association was found in case of coinfection. In further analyses, the anti-GMZ2 IgG concentration on D84 was significantly higher in the S. haematobium-infected and significantly lower in the Strongyloides stercoralis-infected groups, compared to helminth-negative volunteers. Interesting, in the absence of helminth infection, a high anti-GMZ2 IgG concentration on D84 was significantly associated with protection against malaria. Conclusions Our results suggest that helminth infection may reduce naturally acquired and vaccine-induced protection against malaria. Vaccine-specific antibody concentrations on D84 may be associated with protection in participants with no helminth infection. These results suggest that helminth infection affect malaria vaccine immunogenicity and efficacy in helminth endemic countries. Helminths, mainly because of their immune regulatory effects, are able to impact the response induced by vaccines. In the context of clinical trial designs that measure accrual of natural infections during follow up or outcome of controlled human malaria infection (CHMI), their effect on vaccine efficacy can be measured. Indeed, most of such clinical trials on malaria vaccine candidates conducted in Africa, especially where the prevalence of helminths is high, have shown a certain limit in their efficacy and immunogenicity, as compared to results observed in European and U.S volunteers. The present analysis assessed the effect of helminths on GMZ2, a malaria vaccine candidate. We found a high level of anti-GMZ2 antibodies among volunteers not infected with helminths and protected against CHMI, indicating efficacy of the candidate vaccine in this population. We found a species-dependent effect of helminths on the level of post-immunization GMZ2-specific IgG concentration, and an association of helminths with an early onset of malaria in CHMI. Our findings reveal that helminths are associated with immunogenicity and may decrease the protective effect of antibodies induced by vaccination. Helminth infection status shall be determined when measuring the immunogenicity and efficacy of malaria vaccine candidates in helminth endemic countries.
Collapse
Affiliation(s)
- Odilon Nouatin
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany.,Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d'Abomey-Calavi, Cotonou, Bénin
| | | | - Jean Claude Dejon-Agobé
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Amsterdam, The Netherlands
| | - Rolf Fendel
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Javier Ibáñez
- Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany
| | | | | | - Bayodé Roméo Adégbité
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Amsterdam, The Netherlands.,Fondation pour la Recherche Scientifique, Cotonou, Bénin
| | - Yabo Josiane Honkpéhédji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Fondation pour la Recherche Scientifique, Cotonou, Bénin.,Department of Parasitology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Jeannot Fréjus Zinsou
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Fondation pour la Recherche Scientifique, Cotonou, Bénin.,Department of Parasitology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Aurore Bouyoukou Hounkpatin
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany
| | - Kabirou Moutairou
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d'Abomey-Calavi, Cotonou, Bénin
| | - Andreas Homoet
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany
| | - Meral Esen
- Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Andrea Kreidenweiss
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | | | - Michael Theisen
- Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Adrian J F Luty
- Centre d'Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l'Enfance, Calavi, Bénin.,Université de Paris, MERIT, IRD, Paris, France
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Ghyslain Mombo-Ngoma
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany.,Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I, Department of Medicine, University Medical Centre, Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Ramharter
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I, Department of Medicine, University Medical Centre, Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Kremsner
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ayôla Akim Adegnika
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Fondation pour la Recherche Scientifique, Cotonou, Bénin.,Department of Parasitology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| |
Collapse
|
39
|
Lamsfus Calle C, Fendel R, Singh A, Richie TL, Hoffman SL, Kremsner PG, Mordmüller B. Expansion of Functional Myeloid-Derived Suppressor Cells in Controlled Human Malaria Infection. Front Immunol 2021; 12:625712. [PMID: 33815377 PMCID: PMC8017236 DOI: 10.3389/fimmu.2021.625712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Malaria can cause life-threatening complications which are often associated with inflammatory reactions. More subtle, but also contributing to the burden of disease are chronic, often subclinical infections, which result in conditions like anemia and immunologic hyporesponsiveness. Although very frequent, such infections are difficult to study in endemic regions because of interaction with concurrent infections and immune responses. In particular, knowledge about mechanisms of malaria-induced immunosuppression is scarce. We measured circulating immune cells by cytometry in healthy, malaria-naïve, adult volunteers undergoing controlled human malaria infection (CHMI) with a focus on potentially immunosuppressive cells. Infectious Plasmodium falciparum (Pf) sporozoites (SPZ) (PfSPZ Challenge) were inoculated during two independent studies to assess malaria vaccine efficacy. Volunteers were followed daily until parasites were detected in the circulation by RT-qPCR. This allowed us to analyze immune responses during pre-patency and at very low parasite densities in malaria-naïve healthy adults. We observed a consistent increase in circulating polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) in volunteers who developed P. falciparum blood stage parasitemia. The increase was independent of preceding vaccination with a pre-erythrocytic malaria vaccine. PMN-MDSC were functional, they suppressed CD4+ and CD8+ T cell proliferation as shown by ex-vivo co-cultivation with stimulated T cells. PMN-MDSC reduced T cell proliferation upon stimulation by about 50%. Interestingly, high circulating PMN-MDSC numbers were associated with lymphocytopenia. The number of circulating regulatory T cells (Treg) and monocytic MDSC (M-MDSC) showed no significant parasitemia-dependent variation. These results highlight PMN-MDSC in the peripheral circulation as an early indicator of infection during malaria. They suppress CD4+ and CD8+ T cell proliferation in vitro. Their contribution to immunosuppression in vivo in subclinical and uncomplicated malaria will be the subject of further research. Pre-emptive antimalarial pre-treatment of vaccinees to reverse malaria-associated PMN-MDSC immunosuppression could improve vaccine response in exposed individuals.
Collapse
Affiliation(s)
| | - Rolf Fendel
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
| | - Anurag Singh
- Department of Pediatrics 1, University Children's Hospital Tübingen, Tübingen, Germany.,Institute for Clinical and Experimental Transfusion Medicine, University Hospital Tübingen, Tübingen, Germany
| | | | | | - Peter G Kremsner
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
| | - Benjamin Mordmüller
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
| |
Collapse
|
40
|
Abstract
Introduction: An effective vaccine against malaria forms a global health priority. Both naturally acquired immunity and sterile protection induced by irradiated sporozoite immunization were described decades ago. Still no vaccine exists that sufficiently protects children in endemic areas. Identifying immunological correlates of vaccine efficacy can inform rational vaccine design and potentially accelerate clinical development.Areas covered: We discuss recent research on immunological correlates of malaria vaccine efficacy, including: insights from state-of-the-art omics platforms and systems vaccinology analyses; functional anti-parasitic assays; pre-immunization predictors of vaccine efficacy; and comparison of correlates of vaccine efficacy against controlled human malaria infections (CHMI) and against naturally acquired infections.Expert Opinion: Effective vaccination may be achievable without necessarily understanding immunological correlates, but the relatively disappointing efficacy of malaria vaccine candidates in target populations is concerning. Hypothesis-generating omics and systems vaccinology analyses, alongside assessment of pre-immunization correlates, have the potential to bring about paradigm-shifts in malaria vaccinology. Functional assays may represent in vivo effector mechanisms, but have scarcely been formally assessed as correlates. Crucially, evidence is still meager that correlates of vaccine efficacy against CHMI correspond with those against naturally acquired infections in target populations. Finally, the diversity of immunological assays and efficacy endpoints across malaria vaccine trials remains a major confounder.
Collapse
Affiliation(s)
| | - Matthew B B McCall
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
41
|
Ozberk V, Reynolds S, Huo Y, Calcutt A, Eskandari S, Dooley J, Mills JL, Rasmussen IS, Dietrich J, Pandey M, Good MF. Prime-Pull Immunization with a Bivalent M-Protein and Spy-CEP Peptide Vaccine Adjuvanted with CAF®01 Liposomes Induces Both Mucosal and Peripheral Protection from covR/S Mutant Streptococcus pyogenes. mBio 2021; 12:e03537-20. [PMID: 33622722 PMCID: PMC8545125 DOI: 10.1128/mbio.03537-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/15/2021] [Indexed: 11/20/2022] Open
Abstract
Infections with Streptococcus pyogenes and their sequelae are responsible for an estimated 18 million cases of serious disease with >700 million new primary cases and 500,000 deaths per year. Despite the burden of disease, there is currently no vaccine available for this organism. Here, we define a combination vaccine P*17/K4S2 comprising of 20-mer B-cell peptide epitopes, p*17 (a mutant derived from the highly conserved C3-repeat region of the M-protein), and K4S2 (derived from the streptococcal anti-neutrophil factor, Spy-CEP). The peptides are chemically conjugated to either diphtheria toxoid (DT) or a nontoxic mutant form of diphtheria toxin, CRM197. We demonstrate that a prime-pull immunization regimen involving two intramuscular inoculations with P*17/K4S2 adjuvanted with a two-component liposomal adjuvant system (CAF01; developed by Statens Serum Institut [SSI], Denmark), followed by an intranasal inoculation of unadjuvanted vaccine (in Tris) induces peptide- and S. pyogenes-binding antibodies and protects from mucosal and skin infection with hypervirulent covR/S mutant organisms. Prior vaccination with DT does not diminish the response to the conjugate peptide vaccines. Detailed Good Laboratory Practice (GLP) toxicological evaluation in male and female rats did not reveal any gross or histopathological adverse effects.IMPORTANCE A vaccine to control S. pyogenes infection is desperately warranted. S. pyogenes colonizes the upper respiratory tract (URT) and skin, from where it can progress to invasive and immune-mediated diseases. Global mortality estimates for S. pyogenes-associated diseases exceeds 500,000 deaths per year. S. pyogenes utilizes antigenic variation as a defense mechanism to circumvent host immune responses and thus a successful vaccine needs to provide strain-transcending and multicompartment (mucosal and skin) immunity. By defining highly conserved and protective epitopes from two critical virulence factors (M-protein and Spy-CEP) and combining them with a potent immunostimulant, CAF®01, we are addressing an unmet clinical need for a mucosally and skin-active subunit vaccine. We demonstrate that prime-pull immunization (2× intramuscular injections followed by intranasal immunization) promotes high sustained antibody levels in the airway mucosa and serum and protects against URT and invasive disease.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Bacterial/blood
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Bacterial Outer Membrane Proteins/administration & dosage
- Bacterial Outer Membrane Proteins/genetics
- Bacterial Outer Membrane Proteins/immunology
- Epitopes, B-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Female
- Immunity, Mucosal
- Immunization/methods
- Liposomes/administration & dosage
- Liposomes/chemistry
- Male
- Mice, Inbred BALB C
- Rats
- Rats, Sprague-Dawley
- Streptococcal Infections/prevention & control
- Streptococcal Vaccines/administration & dosage
- Streptococcal Vaccines/immunology
- Streptococcus pyogenes/genetics
- Streptococcus pyogenes/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- Mice
Collapse
Affiliation(s)
- Victoria Ozberk
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Simone Reynolds
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Yongbao Huo
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Ainslie Calcutt
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | | | - Jessica Dooley
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Jamie-Lee Mills
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Ida S Rasmussen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Jes Dietrich
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Manisha Pandey
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Michael F Good
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| |
Collapse
|
42
|
Singh SK, Plieskatt J, Chourasia BK, Fabra-García A, Garcia-Senosiain A, Singh V, Bengtsson KL, Reimer JM, Sauerwein R, Jore MM, Theisen M. A Reproducible and Scalable Process for Manufacturing a Pfs48/45 Based Plasmodium falciparum Transmission-Blocking Vaccine. Front Immunol 2021; 11:606266. [PMID: 33505395 PMCID: PMC7832176 DOI: 10.3389/fimmu.2020.606266] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/20/2020] [Indexed: 11/14/2022] Open
Abstract
The cysteine-rich Pfs48/45 protein, a Plasmodium falciparum sexual stage surface protein, has been advancing as a candidate antigen for a transmission-blocking vaccine (TBV) for malaria. However, Pfs48/45 contains multiple disulfide bonds, that are critical for proper folding and induction of transmission-blocking (TB) antibodies. We have previously shown that R0.6C, a fusion of the 6C domain of Pfs48/45 and a fragment of PfGLURP (R0), expressed in Lactococcus lactis, was properly folded and induced transmission-blocking antibodies. Here we describe the process development and technology transfer of a scalable and reproducible process suitable for R0.6C manufacturing under current Good Manufacturing Practices (cGMP). This process resulted in a final purified yield of 25 mg/L, sufficient for clinical evaluation. A panel of analytical assays for release and stability assessment of R0.6C were developed including HPLC, SDS-PAGE, and immunoblotting with the conformation-dependent TB mAb45.1. Intact mass analysis of R0.6C confirmed the identity of the product including the three disulfide bonds and the absence of post-translational modifications. Multi-Angle Light Scattering (MALS) coupled to size exclusion chromatography (SEC-MALS), further confirmed that R0.6C was monomeric (~70 kDa) in solution. Lastly, preclinical studies demonstrated that the R0.6C Drug Product (adsorbed to Alhydrogel®) elicited functional antibodies in small rodents and that adding Matrix-M™ adjuvant further increased the functional response. Here, building upon our past work, we filled the gap between laboratory and manufacturing to ready R0.6C for production under cGMP and eventual clinical evaluation as a malaria TB vaccine.
Collapse
Affiliation(s)
- Susheel K Singh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jordan Plieskatt
- PATH's Malaria Vaccine Initiative, Washington, DC, United States
| | - Bishwanath K Chourasia
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Amanda Fabra-García
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Asier Garcia-Senosiain
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Vandana Singh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Robert Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthijs M Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
43
|
Development of sustainable research excellence with a global perspective on infectious diseases: Centre de Recherches Médicales de Lambaréné (CERMEL), Gabon. Wien Klin Wochenschr 2021; 133:500-508. [PMID: 33398458 PMCID: PMC7781170 DOI: 10.1007/s00508-020-01794-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/30/2020] [Indexed: 11/17/2022]
Abstract
Medical research in sub-Saharan Africa is of high priority for societies to respond adequately to local health needs. Often enough it remains a challenge to build up capacity in infrastructure and human resources to highest international standards and to sustain this over mid-term to long-term periods due to difficulties in obtaining long-term institutional core funding, attracting highly qualified scientists for medical research and coping with ever changing structural and political environments. The Centre de Recherches Médicales de Lambaréné (CERMEL) serves as model for how to overcome such challenges and to continuously increase its impact on medical care in Central Africa and beyond. Starting off as a research annex to the Albert Schweitzer Hospital in Lambaréné, Gabon, it has since then expanded its activities to academic and regulatory clinical trials for drugs, vaccines and diagnostics in the field of malaria, tuberculosis, and a wide range of poverty related and neglected tropical infectious diseases. Advancing bioethics in medical research in Africa and steadily improving its global networks and infrastructures, CERMEL serves as a reference centre for several international consortia. In close collaboration with national authorities, CERMEL has become one of the main training hubs for medical research in Central Africa. It is hoped that CERMEL and its leitmotiv “to improve medical care for local populations” will serve as an inspiration to other institutions in sub-Saharan Africa to further increase African capacity to advance medicine.
Collapse
|
44
|
Ferluga J, Singh I, Rout S, Al-Qahtani A, Yasmin H, Kishore U. Immune Responses in Malaria and Vaccine Strategies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:273-291. [PMID: 34661899 DOI: 10.1007/978-3-030-67452-6_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Malaria is a pandemic with nearly half of global population at risk, caused by parasite Plasmodium species, particularly P. falciparum with a high morbidity and mortality, especially among children. There is an urgent need for development of population protective vaccines, such as in sub-Saharan low-income countries, where P. falciparum malaria is endemic. After years of endeavour with children and adults for safety and efficacy clinical trials, the P. falciparum circumsporozoite protein antigen, is targeted by specific antibodies induced by recombinant vaccine, called TRS,S. TRS,S has been authorized by WHO and Malawi Government to be the first malaria vaccine for up to 2 years of aged children for protection against malaria. Other malaria vaccines in clinical trials are also very promising candidates, including the original live, X-ray attenuated P-sporozoite vaccine, inducing antigen-specific T cell immunity at liver stage. Malaria parasite at blood symptomatic stage is targeted by specific antibodies to parasite-infected erythrocytes, which are important against pathogenic placenta-infected erythrocyte sequestration. Here, the demographic distribution of Plasmodium species and their pathogenicity in infected people are discussed. The role of innate phagocytic cells and malaria antigen specific T cell immunity, as well as that of specific antibody production by B cells are highlighted. The paramount role of cytotoxic CD8+ T cellular immunity in malaria people protection is also included.
Collapse
Affiliation(s)
- Janez Ferluga
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | - Iesha Singh
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sashmita Rout
- Department of Physiology, All-India Institute of Medical Sciences, Bhubaneswar, India
| | - Ahmed Al-Qahtani
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| |
Collapse
|
45
|
Jongo SA, Urbano V, Church LWP, Olotu A, Manock SR, Schindler T, Mtoro A, KC N, Hamad A, Nyakarungu E, Mpina M, Deal A, Bijeri JR, Ondo Mangue ME, Ntutumu Pasialo BE, Nguema GN, Owono SN, Rivas MR, Chemba M, Kassim KR, James ER, Stabler TC, Abebe Y, Saverino E, Sax J, Hosch S, Tumbo AM, Gondwe L, Segura JL, Falla CC, Phiri WP, Hergott DEB, García GA, Schwabe C, Maas CD, Murshedkar T, Billingsley PF, Tanner M, Ayekaba MO, Sim BKL, Daubenberger C, Richie TL, Abdulla S, Hoffman SL. Immunogenicity and Protective Efficacy of Radiation-Attenuated and Chemo-Attenuated PfSPZ Vaccines in Equatoguinean Adults. Am J Trop Med Hyg 2021; 104:283-293. [PMID: 33205741 PMCID: PMC7790068 DOI: 10.4269/ajtmh.20-0435] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/07/2020] [Indexed: 11/15/2022] Open
Abstract
Plasmodium falciparum sporozoite (PfSPZ) Vaccine (radiation-attenuated, aseptic, purified, cryopreserved PfSPZ) and PfSPZ-CVac (infectious, aseptic, purified, cryopreserved PfSPZ administered to subjects taking weekly chloroquine chemoprophylaxis) have shown vaccine efficacies (VEs) of 100% against homologous controlled human malaria infection (CHMI) in nonimmune adults. Plasmodium falciparum sporozoite-CVac has never been assessed against CHMI in African vaccinees. We assessed the safety, immunogenicity, and VE against homologous CHMI of three doses of 2.7 × 106 PfSPZ of PfSPZ Vaccine at 8-week intervals and three doses of 1.0 × 105 PfSPZ of PfSPZ-CVac at 4-week intervals with each arm randomized, double-blind, placebo-controlled, and conducted in parallel. There were no differences in solicited adverse events between vaccinees and normal saline controls, or between PfSPZ Vaccine and PfSPZ-CVac recipients during the 6 days after administration of investigational product. However, from days 7-13, PfSPZ-CVac recipients had significantly more AEs, probably because of Pf parasitemia. Antibody responses were 2.9 times higher in PfSPZ Vaccine recipients than PfSPZ-CVac recipients at time of CHMI. Vaccine efficacy at a median of 14 weeks after last PfSPZ-CVac dose was 55% (8 of 13, P = 0.051) and at a median of 15 weeks after last PfSPZ Vaccine dose was 27% (5 of 15, P = 0.32). The higher VE in PfSPZ-CVac recipients of 55% with a 27-fold lower dose was likely a result of later stage parasite maturation in the liver, leading to induction of cellular immunity against a greater quantity and broader array of antigens.
Collapse
Affiliation(s)
- Said A. Jongo
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Vicente Urbano
- Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | | | - Ally Olotu
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | | | - Ali Mtoro
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Natasha KC
- Sanaria Inc., Rockville, Maryland
- Protein Potential LLC, Rockville, Maryland
| | - Ali Hamad
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Elizabeth Nyakarungu
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | - Anna Deal
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - José Raso Bijeri
- Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | - Martin Eka Ondo Mangue
- Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | | | - Genaro Nsue Nguema
- Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | - Salomon Nguema Owono
- Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | - Matilde Riloha Rivas
- Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | - Mwajuma Chemba
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | - Kamaka R. Kassim
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | | | | | | | - Julian Sax
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Salome Hosch
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | - Linda Gondwe
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - J. Luis Segura
- Medical Care Development International, Silver Spring, Maryland
| | | | | | | | | | | | - Carl D. Maas
- Marathon EG Production, Ltd., Bioko Norte, Equatorial Guinea
| | | | | | - Marcel Tanner
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Mitoha Ondo’o Ayekaba
- Ministry of Health and Social Welfare, Government of Equatorial Guinea, Bioko Norte, Equatorial Guinea
| | - B. Kim Lee Sim
- Sanaria Inc., Rockville, Maryland
- Protein Potential LLC, Rockville, Maryland
| | | | | | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | |
Collapse
|
46
|
de Araújo MV, Santos Júnior SRD, Nosanchuk JD, Taborda CP. Therapeutic Vaccination with Cationic Liposomes Formulated with Dioctadecyldimethylammonium and Trehalose Dibehenate (CAF01) and Peptide P10 Is Protective in Mice Infected with Paracoccidioides brasiliensis. J Fungi (Basel) 2020; 6:jof6040347. [PMID: 33302372 PMCID: PMC7762540 DOI: 10.3390/jof6040347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/25/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
The peptide P10 is a vaccine candidate for Paracoccidioidomycosis, a systemic mycosis caused by fungal species of the genus Paracoccidioides spp. We have previously shown that peptide P10 vaccination, in the presence of several different adjuvants, induced a protective cellular immune response mediated by CD4+ Th1 lymphocytes that was associated with the increased production of IFN-γ in mice challenged with a virulent isolate of Paracoccidoides brasiliensis. Cationic liposomes formulated with dioctadecyldimethylammonium and trehalose dibehenate (DDA/TDB, termed also CAF01–cationic adjuvant formulation) have been developed for safe administration in humans and CAF01 liposomes are utilized as an adjuvant for modulating a robust Th1/Th17 cellular response. We evaluated the efficacy of the adsorption of peptide P10 to CAF01 cationic liposomes and used the generated liposomes to vaccinate C57Bl/6 mice infected with P. brasiliensis. Our results showed that P10 was efficiently adsorbed onto CAF01 liposomes. The vaccination of infected mice with cationic liposomes formulated with DDA/TDB 250/50 µg/mL and 20 µg of P10 induced an effective cellular immune response with increased levels of Th17 cytokines, which correlated with significant decreases in the fungal burdens in lungs and protective granulomatous tissue responses. Hence, cationic liposomes of DDA/TDB 250/50 µg/mL with 20 µg of P10 are a promising therapeutic for safely and effectively improving the treatment of paracoccidioidomycosis.
Collapse
Affiliation(s)
- Marcelo Valdemir de Araújo
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (M.V.d.A.); (S.R.D.S.J.)
| | - Samuel Rodrigues Dos Santos Júnior
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (M.V.d.A.); (S.R.D.S.J.)
| | - Joshua D. Nosanchuk
- Departments of Medicine (Division of Infectious Disease), Microbiology and Immunology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA;
| | - Carlos Pelleschi Taborda
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil; (M.V.d.A.); (S.R.D.S.J.)
- Departamento de Dermatologia, Instituto de Medicina Tropical de São Paulo—LIM53, Faculdade de Medicina, Universidade de São Paulo, São Paulo 4023-062, Brazil
- Correspondence: ; Tel.: +55-11-3091-7351
| |
Collapse
|
47
|
Diversify and Conquer: The Vaccine Escapism of Plasmodium falciparum. Microorganisms 2020; 8:microorganisms8111748. [PMID: 33171746 PMCID: PMC7694999 DOI: 10.3390/microorganisms8111748] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Over the last century, a great deal of effort and resources have been poured into the development of vaccines to protect against malaria, particularly targeting the most widely spread and deadly species of the human-infecting parasites: Plasmodium falciparum. Many of the known proteins the parasite uses to invade human cells have been tested as vaccine candidates. However, precisely because of the importance and immune visibility of these proteins, they tend to be very diverse, and in many cases redundant, which limits their efficacy in vaccine development. With the advent of genomics and constantly improving sequencing technologies, an increasingly clear picture is emerging of the vast genomic diversity of parasites from different geographic areas. This diversity is distributed throughout the genome and includes most of the vaccine candidates tested so far, playing an important role in the low efficacy achieved. Genomics is a powerful tool to search for genes that comply with the most desirable attributes of vaccine targets, allowing us to evaluate function, immunogenicity and also diversity in the worldwide parasite populations. Even predicting how this diversity might evolve and spread in the future becomes possible, and can inform novel vaccine efforts.
Collapse
|
48
|
Achan J, Reuling IJ, Yap XZ, Dabira E, Ahmad A, Cox M, Nwakanma D, Tetteh K, Wu L, Bastiaens GJH, Abebe Y, Manoj A, Kaur H, Miura K, Long C, Billingsley PF, Sim BKL, Hoffman SL, Drakeley C, Bousema T, D’Alessandro U. Serologic Markers of Previous Malaria Exposure and Functional Antibodies Inhibiting Parasite Growth Are Associated With Parasite Kinetics Following a Plasmodium falciparum Controlled Human Infection. Clin Infect Dis 2020; 70:2544-2552. [PMID: 31402382 PMCID: PMC7286377 DOI: 10.1093/cid/ciz740] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/01/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We assessed the impact of exposure to Plasmodium falciparum on parasite kinetics, clinical symptoms, and functional immunity after controlled human malaria infection (CHMI) in 2 cohorts with different levels of previous malarial exposure. METHODS Nine adult males with high (sero-high) and 10 with low (sero-low) previous exposure received 3200 P. falciparum sporozoites (PfSPZ) of PfSPZ Challenge by direct venous inoculation and were followed for 35 days for parasitemia by thick blood smear (TBS) and quantitative polymerase chain reaction. Endpoints were time to parasitemia, adverse events, and immune responses. RESULTS Ten of 10 (100%) volunteers in the sero-low and 7 of 9 (77.8%) in the sero-high group developed parasitemia detected by TBS in the first 28 days (P = .125). The median time to parasitemia was significantly shorter in the sero-low group than the sero-high group (9 days [interquartile range {IQR} 7.5-11.0] vs 11.0 days [IQR 7.5-18.0], respectively; log-rank test, P = .005). Antibody recognition of sporozoites was significantly higher in the sero-high (median, 17.93 [IQR 12.95-24] arbitrary units [AU]) than the sero-low volunteers (median, 10.54 [IQR, 8.36-12.12] AU) (P = .006). Growth inhibitory activity was significantly higher in the sero-high (median, 21.8% [IQR, 8.15%-29.65%]) than in the sero-low group (median, 8.3% [IQR, 5.6%-10.23%]) (P = .025). CONCLUSIONS CHMI was safe and well tolerated in this population. Individuals with serological evidence of higher malaria exposure were able to better control infection and had higher parasite growth inhibitory activity. CLINICAL TRIALS REGISTRATION NCT03496454.
Collapse
Affiliation(s)
- Jane Achan
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Isaie J Reuling
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Xi Zen Yap
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Edgard Dabira
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Abdullahi Ahmad
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Momodou Cox
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Davis Nwakanma
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Kevin Tetteh
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Lindsey Wu
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Guido J H Bastiaens
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Harparkash Kaur
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - Carole Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | | | | | | | - Chris Drakeley
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Umberto D’Alessandro
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| |
Collapse
|
49
|
Nouatin O, Ateba Ngoa U, Ibáñez J, Dejon-Agobe JC, Mordmüller B, Edoa JR, Mougeni F, Brückner S, Bouyoukou Hounkpatin A, Esen M, Theisen M, Moutairou K, Hoffman SL, Issifou S, Luty AJF, Loembe MM, Agnandji ST, Lell B, Kremsner PG, Adegnika AA. Effect of immune regulatory pathways after immunization with GMZ2 malaria vaccine candidate in healthy lifelong malaria-exposed adults. Vaccine 2020; 38:4263-4272. [PMID: 32386747 PMCID: PMC7297038 DOI: 10.1016/j.vaccine.2020.04.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/08/2020] [Accepted: 04/20/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Despite appreciable immunogenicity in malaria-naive populations, many candidate malaria vaccines are considerably less immunogenic in malaria-exposed populations. This could reflect induction of immune regulatory mechanisms involving Human Leukocyte Antigen G (HLA-G), regulatory T (Treg), and regulatory B (Breg) cells. Here, we addressed the question whether there is correlation between these immune regulatory pathways and both plasmablast frequencies and vaccine-specific IgG concentrations. METHODS Fifty Gabonese adults with lifelong exposure to Plasmodium spp were randomized to receive three doses of either 30 µg or 100 µg GMZ2-CAF01, or 100 µg GMZ2-alum, or control vaccine (rabies vaccine) at 4-week intervals. Only plasma and peripheral blood mononuclear cells isolated from blood samples collected before (D0) and 28 days after the third vaccination (D84) of 35 participants were used to measure sHLA-G levels and anti-GMZ2 IgG concentrations, and to quantify Treg, Breg and plasmablast cells. Vaccine efficacy was assessed using controlled human malaria infection (CHMI) by direct venous inoculation of Plasmodium falciparum sporozoites (PfSPZ Challenge). RESULTS The sHLA-G concentration increased from D0 to D84 in all GMZ2 vaccinated participants and in the control group, whereas Treg frequencies increased only in those receiving 30 µg or 100 µg GMZ2-CAF01. The sHLA-G level on D84 was associated with a decrease of the anti-GMZ2 IgG concentration, whereas Treg frequencies on D0 or on D84, and Breg frequency on D84 were associated with lower plasmablast frequencies. Importantly, having a D84:D0 ratio of sHLA-G above the median was associated with an increased risk of P. falciparum infection after sporozoites injection. CONCLUSION Regulatory immune responses are induced following immunization. Stronger sHLA-G and Treg immune responses may suppress vaccine induced immune responses, and the magnitude of the sHLA-G response increased the risk of Plasmodium falciparum infection after CHMI. These findings could have implications for the design and testing of malaria vaccine candidates in semi-immune individuals.
Collapse
Affiliation(s)
- Odilon Nouatin
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany; Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d'Abomey-Calavi, Cotonou, Benin.
| | - Ulysse Ateba Ngoa
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon.
| | - Javier Ibáñez
- Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany
| | - Jean Claude Dejon-Agobe
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany.
| | - Jean Ronald Edoa
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany.
| | - Fabrice Mougeni
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon
| | - Sina Brückner
- Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany.
| | - Aurore Bouyoukou Hounkpatin
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany
| | - Meral Esen
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany.
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark and Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark.
| | - Kabirou Moutairou
- Département de Biochimie et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d'Abomey-Calavi, Cotonou, Benin.
| | | | - Saadou Issifou
- Fondation pour la Recherche Scientifique, 72 BP45 Cotonou, Benin.
| | - Adrian J F Luty
- Centre d'Etude et de Recherche sur le Paludisme Associé à la Grossesse et à l'Enfance, Faculté des Sciences de la Santé, Université d'Abomey-Calavi, Cotonou, MERIT UMR D216, Benin; Université de Paris, MERIT, IRD, Paris, France.
| | - Marguerite M Loembe
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany.
| | - Selidji Todagbé Agnandji
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany.
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria.
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany.
| | - Ayôla Akim Adegnika
- Centre de Recherches Médicales de Lambaréné, BP: 242 Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tubingen, Wilhelmstraβe 27, D-72074 Tübingen, Germany; Germany; German Centre for Infection Research (DZIF), Partner Site Tübingen, Germany; Leiden University Medical Centre (LUMC), 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
50
|
Schmaler M, Orlova-Fink N, Rutishauser T, Abdulla S, Daubenberger C. Human unconventional T cells in Plasmodium falciparum infection. Semin Immunopathol 2020; 42:265-277. [PMID: 32076813 PMCID: PMC7223888 DOI: 10.1007/s00281-020-00791-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 02/07/2020] [Indexed: 12/22/2022]
Abstract
Malaria is an old scourge of humankind and has a large negative impact on the economic development of affected communities. Recent success in malaria control and reduction of mortality seems to have stalled emphasizing that our current intervention tools need to be complemented by malaria vaccines. Different populations of unconventional T cells such as mucosal-associated invariant T (MAIT) cells, invariant natural killer T (iNKT) cells and γδ T cells are gaining attention in the field of malaria immunology. Significant advances in our basic understanding of unconventional T cell biology in rodent malaria models have been made, however, their roles in humans during malaria are less clear. Unconventional T cells are abundant in skin, gut and liver tissues, and long-lasting expansions and functional alterations were observed upon malaria infection in malaria naïve and malaria pre-exposed volunteers. Here, we review the current understanding of involvement of unconventional T cells in anti-Plasmodium falciparum immunity and highlight potential future research avenues.
Collapse
Affiliation(s)
- Mathias Schmaler
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Nina Orlova-Fink
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Tobias Rutishauser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland
| | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Clinical Trial Unit, Bagamoyo, Tanzania
| | - Claudia Daubenberger
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, University of Basel, Basel, Switzerland.
| |
Collapse
|