1
|
Venkatraman N, Tiono AB, Bowyer G, Bellamy DG, Stockdale LK, Powlson J, Collins KA, Coulibaly S, Datoo MS, Silman D, Ouedraogo A, Nébié I, Imoukhuede EB, Brod F, Folegatti P, Dickinson-Craig E, Jamieson S, Bougouma EC, Wright D, Diarra A, Bliss CM, Morter R, Glenn G, Fries LF, Reimer JM, Lövgren-Bengtsson K, Baker M, Poulton I, Moyle S, Berrie E, Green N, Mukhopadhyay E, Viebig NK, Angus B, Lawrie A, Roberts R, Gilbert SC, Lewis DJM, Sirima SB, Ewer KJ, Hill AVS. Evaluation of a novel malaria anti-sporozoite vaccine candidate, R21 in Matrix-M adjuvant, in the UK and Burkina Faso: two phase 1, first-in-human trials. THE LANCET. MICROBE 2025; 6:100868. [PMID: 39805302 DOI: 10.1016/s2666-5247(24)00084-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/08/2024] [Accepted: 03/20/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Malaria remains a substantial public health burden among young children in sub-Saharan Africa and a highly efficacious vaccine eliciting a durable immune response would be a useful tool for controlling malaria. R21 is a malaria vaccine comprising nanoparticles, formed from a circumsporozoite protein and hepatitis B surface antigen (HBsAg) fusion protein, without any unfused HBsAg, and is administered with the saponin-based Matrix-M adjuvant. This study aimed to assess the safety and immunogenicity of the malaria vaccine candidate, R21, administered with or without adjuvant Matrix-M in adults naïve to malaria infection and in healthy adults from malaria endemic areas. METHODS In this Article we report two phase 1, first-in-human trials. The first trial was a phase 1a open-label study in the UK evaluating the safety and immunogenicity of R21 administered either alone, or with 50 μg of Matrix-M. The second trial was a phase 1b randomised controlled trial in Burkina Faso. Adults had to be aged 18-50 years for enrolment in the phase 1a trial, and 18-45 years in the phase 1b trial. The phase 1a trial doses were 2 μg, 10 μg, and 50 μg R21/Matrix-M, and 50 μg R21 only. The phase 1b trial doses were 10 μg R21/Matrix-M and saline placebo. Matrix-M was always dosed at 50 μg. Phase 1b implemented block randomisation by randomisation into study groups by an independent statistician based at the University of Oxford using a randomisation code list with allocation concealment using opaque sealed envelopes. The primary objective of the phase 1a trial was to assess the safety and tolerability of R21 with and without Matrix-M. The primary objective of the phase 1b trial was to assess the safety and tolerability of R21 with Matrix-M. Both trials are registered with ClinicalTrials.gov, NCT02572388 for phase 1a and NCT02925403 for phase 1b, and are completed. FINDINGS Between Oct 1, 2015, and Jan 3, 2017, 31 individuals were enrolled in the phase 1a study. Six individuals were assigned to receive 2 μg R21/Matrix-M, 11 to 10 μg R21/Matrix-M, ten to 50 μg R21/Matrix-M, and four to 50 μg R21 only. Between Aug 26, 2016, and Sept 28, 2017, 13 individuals were enrolled in the phase 1b study. Eight individuals were assigned to receive 10 μg R21/Matrix-M, and five to placebo. Vaccinations were well tolerated, and most local and systemic adverse events were mild. There were no serious adverse events deemed related to vaccination. Two serious adverse events occurred. The first in the 10 μg R21/Matrix-M group was worsening of previously undisclosed or undiagnosed palindromic rheumatism and was deemed unlikely to be related to vaccination and the second in the 2 μg R21/Matrix-M was hospital admission for an unplanned excision of a pre-existing Bartholin's cyst, also unrelated to vaccination. In the phase 1a study, a total of 21 adverse events were recorded in the 2 μg R21/Matrix-M group, 103 in the 10 μg R21/Matrix-M group, 94 in the 50 μg R21/Matrix-M group, and 21 in the 50 μg R21 alone group. In the phase 1b study, twelve adverse events were recorded in the 10 μg R21/Matrix-M group and 0 in the placebo group. INTERPRETATION R21 with Matrix-M adjuvant has an acceptable safety profile. These data have formed the basis for efficacy testing of this vaccine. FUNDING The European Commission Framework 7 and The European & Developing Countries Clinical Trials Partnership.
Collapse
Affiliation(s)
- Navin Venkatraman
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Alfred B Tiono
- Centre National de Recherche et Formation sur le Paludisme (CNRFP) Research Unit, Banfora, Burkina Faso
| | - Georgina Bowyer
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Duncan G Bellamy
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Lisa K Stockdale
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Jonathan Powlson
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Katharine A Collins
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Sam Coulibaly
- Centre National de Recherche et Formation sur le Paludisme (CNRFP) Research Unit, Banfora, Burkina Faso
| | - Mehreen S Datoo
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Daniel Silman
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Alphonse Ouedraogo
- Centre National de Recherche et Formation sur le Paludisme (CNRFP) Research Unit, Banfora, Burkina Faso
| | - Issa Nébié
- Centre National de Recherche et Formation sur le Paludisme (CNRFP) Research Unit, Banfora, Burkina Faso
| | - Egeruan B Imoukhuede
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Florian Brod
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Pedro Folegatti
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Emma Dickinson-Craig
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Sophie Jamieson
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Edith C Bougouma
- Centre National de Recherche et Formation sur le Paludisme (CNRFP) Research Unit, Banfora, Burkina Faso
| | - Daniel Wright
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Amidou Diarra
- Centre National de Recherche et Formation sur le Paludisme (CNRFP) Research Unit, Banfora, Burkina Faso
| | - Carly M Bliss
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Richard Morter
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | | | | | | | | | - Megan Baker
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Ian Poulton
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Sarah Moyle
- Clinical Biomanufacturing Facility, University of Oxford, Churchill Hospital, Oxford, UK
| | - Eleanor Berrie
- Clinical Biomanufacturing Facility, University of Oxford, Churchill Hospital, Oxford, UK
| | - Nicola Green
- Clinical Biomanufacturing Facility, University of Oxford, Churchill Hospital, Oxford, UK
| | - Ekta Mukhopadhyay
- Clinical Biomanufacturing Facility, University of Oxford, Churchill Hospital, Oxford, UK
| | - Nicola K Viebig
- European Vaccine Initiative, Universitäts Klinikum Heidelberg, Heidelberg, Germany
| | - Brian Angus
- Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Alison Lawrie
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Rachel Roberts
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Sarah C Gilbert
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - David J M Lewis
- NIHR Imperial Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, London, UK
| | - Sodiomon B Sirima
- Centre National de Recherche et Formation sur le Paludisme (CNRFP) Research Unit, Banfora, Burkina Faso; Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Katie J Ewer
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Adrian V S Hill
- Jenner Institute, University of Oxford-NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
2
|
Ali MS, Stockdale L, Sagara I, Zongo I, Yerbanga RS, Mahamar A, Nikièma F, Tapily A, Sompougdou F, Diarra M, Bellamy D, Provstgaard-Morys S, Zoungrana C, Issiaka D, Haro A, Sanogo K, Sienou AA, Kaya M, Traore S, Dicko OM, Kone Y, Yalcouye H, Thera I, Diarra K, Snell P, Ofori-Anyinam O, Ockenhouse C, Lee C, Ewer K, Tinto H, Djimde A, Ouedraogo JB, Dicko A, Chandramohan D, Greenwood B. The anti-circumsporozoite antibody response to repeated, seasonal booster doses of the malaria vaccine RTS,S/AS01 E. NPJ Vaccines 2025; 10:26. [PMID: 39915506 PMCID: PMC11802723 DOI: 10.1038/s41541-025-01078-0] [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: 09/25/2024] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
The recently deployed RTS,S/AS01E malaria vaccine induces a strong antibody response to the circumsporozoite protein (CSP) on the surface of the Plasmodium falciparum sporozoite which is associated with protection. The anti-CSP antibody titre falls rapidly after primary vaccination, associated with a decline in efficacy, but the antibody titre and the protective response can be partially restored by a booster dose of vaccine, but this response is also transitory. In many malaria- endemic areas of Africa, children are at risk of malaria, including severe malaria, until they are five years of age or older and to sustain protection from malaria for this period by vaccination with RTS,S/AS01E, repeated booster doses of vaccine may be required. However, there is little information about the immune response to repeated booster doses of RTS,S/AS01E. In many malaria-endemic areas of Africa, the burden of malaria is largely restricted to the rainy season and, therefore, a recent trial conducted in Burkina Faso and Mali explored the impact of repeated annual booster doses of RTS,S/AS01E given immediately prior to the malaria transmission season until children reached the age of five years. Anti-CSP antibody titres were measured in sera obtained from a randomly selected subset of children enrolled in this trial collected before and one month after three priming and four annual booster doses of vaccine using the GSK ELISA developed at the University of Ghent and, in a subset of these samples, by a multiplex assay developed at the University of Oxford. Three priming doses of RTS,S/AS01E induced a strong anti-CSP antibody response (GMT 368.9 IU/mL). Subsequent annual, seasonal booster doses induced a strong, but lower, antibody response; the GMT after the fourth booster was 128.5 IU/mL. Children whose antibody response was in the upper and middle terciles post vaccination had a lower incidence of malaria during the following year than children in the lowest tercile. Results obtained with GSK ELISA and the Oxford Multiplex assay were strongly correlated (Pearson's correlation coefficient, r = 0.94; 95% CI, 0.93-0.95). Although anti-CSP antibody titres declined after repeated booster doses of RTS,S/AS01E a high, although declining, level of efficacy was sustained suggesting that there may have been changes in the characteristics of the anti-CSP antibody following repeated booster doses.Clinical Trials Registration. NCT03143218.
Collapse
Affiliation(s)
- M Sanni Ali
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - Issaka Sagara
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Issaka Zongo
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Rakiswendé Serge Yerbanga
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
- Institut des Sciences et Techniques, Bobo-Dioulasso, Burkina Faso
| | - Almahamoudou Mahamar
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Frédéric Nikièma
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Amadou Tapily
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | | | - Modibo Diarra
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | | | | | - Charles Zoungrana
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Djibrilla Issiaka
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Alassane Haro
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
- Institut des Sciences et Techniques, Bobo-Dioulasso, Burkina Faso
| | - Koualy Sanogo
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Abdoul Aziz Sienou
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
- Institut des Sciences et Techniques, Bobo-Dioulasso, Burkina Faso
| | - Mahamadou Kaya
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Seydou Traore
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Oumar M Dicko
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Youssouf Kone
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Hama Yalcouye
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Ismaila Thera
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Kalifa Diarra
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Paul Snell
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | | | | | - Katie Ewer
- GSK Vaccines Institute for Global Health, Sienna, Italy
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Abdoulaye Djimde
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Jean-Bosco Ouedraogo
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
- Institut des Sciences et Techniques, Bobo-Dioulasso, Burkina Faso
| | - Alassane Dicko
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | | | - Brian Greenwood
- London School of Hygiene & Tropical Medicine, London, United Kingdom.
| |
Collapse
|
3
|
Lin L, Spreng RL, Seaton KE, Dennison SM, Dahora LC, Schuster DJ, Sawant S, Gilbert PB, Fong Y, Kisalu N, Pollard AJ, Tomaras GD, Li J. GeM-LR: Discovering predictive biomarkers for small datasets in vaccine studies. PLoS Comput Biol 2024; 20:e1012581. [PMID: 39541411 PMCID: PMC11594404 DOI: 10.1371/journal.pcbi.1012581] [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: 02/05/2024] [Revised: 11/26/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Despite significant progress in vaccine research, the level of protection provided by vaccination can vary significantly across individuals. As a result, understanding immunologic variation across individuals in response to vaccination is important for developing next-generation efficacious vaccines. Accurate outcome prediction and identification of predictive biomarkers would represent a significant step towards this goal. Moreover, in early phase vaccine clinical trials, small datasets are prevalent, raising the need and challenge of building a robust and explainable prediction model that can reveal heterogeneity in small datasets. We propose a new model named Generative Mixture of Logistic Regression (GeM-LR), which combines characteristics of both a generative and a discriminative model. In addition, we propose a set of model selection strategies to enhance the robustness and interpretability of the model. GeM-LR extends a linear classifier to a non-linear classifier without losing interpretability and empowers the notion of predictive clustering for characterizing data heterogeneity in connection with the outcome variable. We demonstrate the strengths and utility of GeM-LR by applying it to data from several studies. GeM-LR achieves better prediction results than other popular methods while providing interpretations at different levels.
Collapse
Affiliation(s)
- Lin Lin
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, United States of America
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, United States of America
| | - Rachel L. Spreng
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, United States of America
| | - Kelly E. Seaton
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, United States of America
- Department of Surgery, Duke University, Durham, North Carolina, United States of America
| | - S. Moses Dennison
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, United States of America
- Department of Surgery, Duke University, Durham, North Carolina, United States of America
| | - Lindsay C. Dahora
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, United States of America
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, United States of America
| | - Daniel J. Schuster
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, United States of America
- Department of Surgery, Duke University, Durham, North Carolina, United States of America
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, United States of America
| | - Sheetal Sawant
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, United States of America
- Department of Surgery, Duke University, Durham, North Carolina, United States of America
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Neville Kisalu
- Center for Vaccine Innovation and Access, PATH, Washington, DC, United States of America
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Georgia D. Tomaras
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, United States of America
- Department of Surgery, Duke University, Durham, North Carolina, United States of America
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, United States of America
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, United States of America
| | - Jia Li
- Department of Statistics, The Pennsylvania State University, Pennsylvania, United States of America
| |
Collapse
|
4
|
Spreng RL, Seaton KE, Lin L, Hilliard S, Horn GQ, Abraha M, Deal AW, Li K, Carnacchi AJ, Feeney E, Shabbir S, Zhang L, Bekker V, Mudrak SV, Dutta S, Mercer LD, Gregory S, King CR, Wille-Reece U, Jongert E, Kisalu NK, Tomaras GD, Dennison SM. Identification of RTS,S/AS01 vaccine-induced humoral biomarkers predictive of protection against controlled human malaria infection. JCI Insight 2024; 9:e178801. [PMID: 39377226 PMCID: PMC11466194 DOI: 10.1172/jci.insight.178801] [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: 01/21/2024] [Accepted: 08/23/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUNDThe mechanism(s) responsible for the efficacy of WHO-recommended malaria vaccine RTS,S/AS01 are not completely understood. We previously identified RTS,S vaccine-induced Plasmodium falciparum circumsporozoite protein-specific (PfCSP-specific) antibody measures associated with protection from controlled human malaria infection (CHMI). Here, we tested the protection-predicting capability of these measures in independent CHMI studies.METHODSVaccine-induced total serum antibody (immunoglobulins, Igs) and subclass antibody (IgG1 and IgG3) responses were measured by biolayer interferometry and the binding antibody multiplex assay, respectively. Immune responses were compared between protected and nonprotected vaccinees using univariate and multivariate logistic regression.RESULTSBlinded prediction analysis showed that 5 antibody binding measures, including magnitude-avidity composite of serum Ig specific for PfCSP, major NANP repeats and N-terminal junction, and PfCSP- and NANP-specific IgG1 subclass magnitude, had good prediction accuracy (area under the receiver operating characteristic curves [ROC AUC] > 0.7) in at least 1 trial. Furthermore, univariate analysis showed a significant association between these antibody measures and protection (odds ratios 2.6-3.1). Multivariate modeling of combined data from 3 RTS,S CHMI trials identified the combination of IgG1 NANP binding magnitude plus serum NANP and N-junction Ig binding magnitude-avidity composite as the best predictor of protection (95% confidence interval for ROC AUC 0.693-0.834).CONCLUSIONThese results reinforce our previous findings and provide a tool for predicting protection in future trials.TRIAL REGISTRATIONClinicalTrials.gov NCT03162614, NCT03824236, NCT01366534, and NCT01857869.FUNDINGThis study was supported by Bill & Melinda Gates Foundation's Global Health-Discovery Collaboratory grants (INV-008612 and INV-043419) to GDT.
Collapse
Affiliation(s)
| | - Kelly E. Seaton
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Lin Lin
- Center for Human Systems Immunology
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | | | - Gillian Q. Horn
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Milite Abraha
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Aaron W. Deal
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Kan Li
- Center for Human Systems Immunology
- Department of Surgery, and
| | | | | | - Siam Shabbir
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Lu Zhang
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Valerie Bekker
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Sarah V. Mudrak
- Center for Human Systems Immunology
- Department of Surgery, and
| | - Sheetij Dutta
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Laina D. Mercer
- Center for Vaccine Innovation and Access, PATH, Seattle, Washington, USA
| | - Scott Gregory
- Center for Vaccine Innovation and Access, PATH, Washington, DC, USA
| | - C. Richter King
- Center for Vaccine Innovation and Access, PATH, Washington, DC, USA
| | | | | | | | - Georgia D. Tomaras
- Duke Human Vaccine Institute
- Center for Human Systems Immunology
- Department of Surgery, and
- Department of Integrative Immunobiology and
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | | |
Collapse
|
5
|
Cummings JF, Polhemus ME, Kester KE, Ockenhouse CF, Gasser RA, Coyne P, Wortmann G, Nielsen RK, Schaecher K, Holland CA, Krzych U, Tornieporth N, Soisson LA, Angov E, Heppner DG. A phase IIa, randomized, double-blind, safety, immunogenicity and efficacy trial of Plasmodium falciparum vaccine antigens merozoite surface protein 1 and RTS,S formulated with AS02 adjuvant in healthy, malaria-naïve adults. Vaccine 2024; 42:3066-3074. [PMID: 38584058 DOI: 10.1016/j.vaccine.2024.03.072] [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: 06/13/2023] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND To improve the efficacy of Plasmodium falciparum malaria vaccine RTS,S/AS02, we conducted a study in 2001 in healthy, malaria-naïve adults administered RTS,S/AS02 in combination with FMP1, a recombinant merozoite surface-protein-1, C-terminal 42kD fragment. METHODS A double-blind Phase I/IIa study randomized N = 60 subjects 1:1:1:1 to one of four groups, N = 15/group, to evaluate safety, immunogenicity, and efficacy of intra-deltoid half-doses of RTS,S/AS02 and FMP1/AS02 administered in the contralateral (RTS,S + FMP1-separate) or same (RTS,S + FMP1-same) sites, or FMP1/AS02 alone (FMP1-alone), or RTS,S/AS02 alone (RTS,S-alone) on a 0-, 1-, 3-month schedule. Subjects receiving three doses of vaccine and non-immunized controls (N = 11) were infected with homologous P. falciparum 3D7 sporozoites by Controlled Human Malaria Infection (CHMI). RESULTS Subjects in all vaccination groups experienced mostly mild or moderate local and general adverse events that resolved within eight days. Anti-circumsporozoite antibody levels were lower when FMP1 and RTS,S were co-administered at the same site (35.0 µg/mL: 95 % CI 20.3-63), versus separate arms (57.4 µg/mL: 95 % CI 32.3-102) or RTS,S alone (62.0 µg/mL: 95 % CI: 37.8-101.8). RTS,S-specific lymphoproliferative responses and ex vivo ELISpot CSP-specific interferon-gamma (IFN-γ) responses were indistinguishable among groups receiving RTS,S/AS02. There was no difference in antibody to FMP1 among groups receiving FMP1/AS02. After CHMI, groups immunized with a RTS,S-containing regimen had ∼ 30 % sterile protection against parasitemia, and equivalent delays in time-to-parasitemia. The FMP1/AS02 alone group showed no sterile immunity or delay in parasitemia. CONCLUSION Co-administration of RTS,S and FMP1/AS02 reduced anti-RTS,S antibody, but did not affect tolerability, cellular immunity, or efficacy in a stringent CHMI model. Absence of efficacy or delay of patency in the sporozoite challenge model in the FMP1/AS02 group did not rule out efficacy of FMP1/AS02 in an endemic population. However, a Phase IIb trial of FMP1/AS02 in children in malaria-endemic Kenya did not demonstrate efficacy against natural infection. CLINICALTRIALS gov identifier: NCT01556945.
Collapse
Affiliation(s)
- J F Cummings
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - M E Polhemus
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - K E Kester
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - C F Ockenhouse
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - R A Gasser
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - P Coyne
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - G Wortmann
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - R K Nielsen
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - K Schaecher
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - C A Holland
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - U Krzych
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | - L A Soisson
- Malaria Vaccine Development Program, United States Agency for International Development, Washington, DC, USA
| | - E Angov
- Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | - D G Heppner
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
6
|
Dobbs KR, Atieli HE, Valim C, Beeson JG. Previous Malaria Exposures and Immune Dysregulation: Developing Strategies To Improve Malaria Vaccine Efficacy in Young Children. Am J Trop Med Hyg 2024; 110:627-630. [PMID: 38442424 PMCID: PMC10993830 DOI: 10.4269/ajtmh.23-0696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/06/2023] [Indexed: 03/07/2024] Open
Abstract
After several decades in development, two malaria vaccines based on the same antigen and with very similar constructs and adjuvants, RTS,S/AS01 (RTS,S) and R21/Matrix-M (R21), were recommended by the WHO for widespread vaccination of children. These vaccines are much-needed additions to malaria control programs that, when used in conjunction with other control measures, will help to accelerate reductions in malaria morbidity and mortality. Although R21 is not yet available, RTS,S is currently being integrated into routine vaccine schedules in some areas. However, the efficacy of RTS,S is partial, short-lived, and varies widely according to age and geographic location. It is not clear why RTS,S induces protection in some individuals and not others, what the immune mechanisms are that favor protective immunity with RTS,S, and how immune mechanisms are influenced by host and environmental factors. Several studies suggest that higher levels of previous malaria exposure negatively impact RTS,S clinical efficacy. In this article, we summarize data suggesting that previous malaria exposures negatively impact the efficacy of RTS,S and other malaria vaccine candidates. We highlight recent evidence suggesting that increasing malaria exposure impairs the generation of functional antibody responses to RTS,S. Finally, we discuss how investigation of clinical and immune factors associated with suboptimal responses to RTS,S can be used to develop strategies to optimize RTS,S, which will remain relevant to R21 and next-generation vaccines.
Collapse
Affiliation(s)
| | | | - Clarissa Valim
- Boston University School of Public Health, Boston, Massachusetts
| | | |
Collapse
|
7
|
Dong Y, Gottardo R. An approach for integrating multimodal omics data into sparse and interpretable models. CELL REPORTS METHODS 2024; 4:100718. [PMID: 38412832 PMCID: PMC10921032 DOI: 10.1016/j.crmeth.2024.100718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/29/2024]
Abstract
Using omics data, a common goal is to identify a concise set of variables that predict a clinical endpoint from an extensive pool. In a recent paper published in Nature Biotechnology, Hédou et al.1 introduced Stabl, a computational method crafted to identify sparse yet robust signatures linked to endpoints.
Collapse
Affiliation(s)
- Yixing Dong
- Lausanne University Hospital and University of Lausanne, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Raphael Gottardo
- Lausanne University Hospital and University of Lausanne, Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
8
|
Locke E, Flores-Garcia Y, Mayer BT, MacGill RS, Borate B, Salgado-Jimenez B, Gerber MW, Mathis-Torres S, Shapiro S, King CR, Zavala F. Establishing RTS,S/AS01 as a benchmark for comparison to next-generation malaria vaccines in a mouse model. NPJ Vaccines 2024; 9:29. [PMID: 38341502 DOI: 10.1038/s41541-024-00819-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
New strategies are needed to reduce the incidence of malaria, and promising approaches include vaccines targeting the circumsporozoite protein (CSP). To improve upon the malaria vaccine, RTS,S/AS01, it is essential to standardize preclinical assays to measure the potency of next-generation vaccines against this benchmark. We focus on RTS,S/AS01-induced antibody responses and functional activity in conjunction with robust statistical analyses. Transgenic Plasmodium berghei sporozoites containing full-length P. falciparum CSP (tgPb-PfCSP) allow two assessments of efficacy: quantitative reduction in liver infection following intravenous challenge, and sterile protection from mosquito bite challenge. Two or three doses of RTS,S/AS01 were given intramuscularly at 3-week intervals, with challenge 2-weeks after the last vaccination. Minimal inter- and intra-assay variability indicates the reproducibility of the methods. Importantly, the range of this model is suitable for screening more potent vaccines. Levels of induced anti-CSP antibody 2A10 equivalency were also associated with activity: 105 μg/mL (95% CI: 68.8, 141) reduced liver infection by 50%, whereas 285 μg/mL (95% CI: 166, 404) is required for 50% sterile protection from mosquito bite challenge. Additionally, the liver burden model was able to differentiate between protected and non-protected human plasma samples from a controlled human malaria infection study, supporting these models' relevance and predictive capability. Comparison in animal models of CSP-based vaccine candidates to RTS,S/AS01 is now possible under well controlled conditions. Assessment of the quality of induced antibodies, likely a determinant of durability of protection in humans, should be possible using these methods.
Collapse
Affiliation(s)
- Emily Locke
- Center for Vaccine Innovation and Access, PATH, Washington, DC, 20001, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Bryan T Mayer
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Randall S MacGill
- Center for Vaccine Innovation and Access, PATH, Washington, DC, 20001, USA
| | - Bhavesh Borate
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Berenice Salgado-Jimenez
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Monica W Gerber
- Vaccine and Infectious Disease Division, Fred Hutchison Cancer Research Center, Seattle, WA, 98109, USA
| | - Shamika Mathis-Torres
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sarah Shapiro
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - C Richter King
- Center for Vaccine Innovation and Access, PATH, Washington, DC, 20001, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
9
|
Roman F, Burny W, Ceregido MA, Laupèze B, Temmerman ST, Warter L, Coccia M. Adjuvant system AS01: from mode of action to effective vaccines. Expert Rev Vaccines 2024; 23:715-729. [PMID: 39042099 DOI: 10.1080/14760584.2024.2382725] [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: 04/17/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION The use of novel adjuvants in human vaccines continues to expand as their contribution to preventing disease in challenging populations and caused by complex pathogens is increasingly understood. AS01 is a family of liposome-based vaccine Adjuvant Systems containing two immunostimulants: 3-O-desacyl-4'-monophosphoryl lipid A and the saponin QS-21. AS01-containing vaccines have been approved and administered to millions of individuals worldwide. AREAS COVERED Here, we report advances in our understanding of the mode of action of AS01 that contributed to the development of efficacious vaccines preventing disease due to malaria, herpes zoster, and respiratory syncytial virus. AS01 induces early innate immune activation that induces T cell-mediated and antibody-mediated responses with optimized functional characteristics and induction of immune memory. AS01-containing vaccines appear relatively impervious to baseline immune status translating into high efficacy across populations. Currently licensed AS01-containing vaccines have shown acceptable safety profiles in clinical trials and post-marketing settings. EXPERT OPINION Initial expectations that adjuvantation with AS01 could support effective vaccine responses and contribute to disease control have been realized. Investigation of the utility of AS01 in vaccines to prevent other challenging diseases, such as tuberculosis, is ongoing, together with efforts to fully define its mechanisms of action in different vaccine settings.
Collapse
|
10
|
Bolton JS, MacGill RS, Locke E, Regules JA, Bergmann-Leitner ES. Novel antibody competition binding assay identifies distinct serological profiles associated with protection. Front Immunol 2023; 14:1303446. [PMID: 38152401 PMCID: PMC10752609 DOI: 10.3389/fimmu.2023.1303446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/21/2023] [Indexed: 12/29/2023] Open
Abstract
Introduction Pre-erythrocytic malaria vaccines hold the promise of inducing sterile protection thereby preventing the morbidity and mortality associated with Plasmodium infection. The main surface antigen of P. falciparum sporozoites, i.e., the circumsporozoite protein (CSP), has been extensively explored as a target of such vaccines with significant success in recent years. Systematic adjuvant selection, refinements of the immunization regimen, and physical properties of the antigen may all contribute to the potential of increasing the efficacy of CSP-based vaccines. Protection appears to be dependent in large part on CSP antibodies. However due to a knowledge gap related to the exact correlates of immunity, there is a critical need to improve our ability to down select candidates preclinically before entering clinical trials including with controlled human malaria infections (CHMI). Methods We developed a novel multiplex competition assay based on well-characterized monoclonal antibodies (mAbs) that target crucial epitopes across the CSP molecule. This new tool assesses both, quality and epitope-specific concentrations of vaccine-induced antibodies by measuring their equivalency with a panel of well-characterized, CSP-epitope-specific mAbs. Results Applying this method to RTS,S-immune sera from a CHMI trial demonstrated a quantitative epitope-specificity profile of antibody responses that can differentiate between protected vs. nonprotected individuals. Aligning vaccine efficacy with quantitation of the epitope fine specificity results of this equivalency assay reveals the importance of epitope specificity. Discussion The newly developed serological equivalence assay will inform future vaccine design and possibly even adjuvant selection. This methodology can be adapted to other antigens and disease models, when a panel of relevant mAbs exists, and could offer a unique tool for comparing and down-selecting vaccine formulations.
Collapse
Affiliation(s)
- Jessica S. Bolton
- Biologics Research & Development, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, United States
| | - Randall S. MacGill
- Center for Vaccine Innovation and Access, PATH, Washington, DC, United States
| | - Emily Locke
- Center for Vaccine Innovation and Access, PATH, Washington, DC, United States
| | - Jason A. Regules
- Biologics Research & Development, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, United States
| | - Elke S. Bergmann-Leitner
- Biologics Research & Development, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, United States
| |
Collapse
|
11
|
Sagara I, Zongo I, Cairns M, Yerbanga RS, Mahamar A, Nikièma F, Tapily A, Sompougdou F, Diarra M, Zoungrana C, Issiaka D, Haro A, Sanogo K, Aziz Sienou A, Kaya M, Traore S, Thera I, Diarra K, Dolo A, Kuepfer I, Snell P, Milligan P, Ockenhouse C, Ofori-Anyinam O, Tinto H, Djimde A, Ouedraogo JB, Dicko A, Chandramohan D, Greenwood B. The Anti-Circumsporozoite Antibody Response of Children to Seasonal Vaccination With the RTS,S/AS01E Malaria Vaccine. Clin Infect Dis 2022; 75:613-622. [PMID: 34894221 PMCID: PMC9464075 DOI: 10.1093/cid/ciab1017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND A trial in African children showed that combining seasonal vaccination with the RTS,S/AS01E vaccine with seasonal malaria chemoprevention reduced the incidence of uncomplicated and severe malaria compared with either intervention given alone. Here, we report on the anti-circumsporozoite antibody response to seasonal RTS,S/AS01E vaccination in children in this trial. METHODS Sera from a randomly selected subset of children collected before and 1 month after 3 priming doses of RTS,S/AS01E and before and 1 month after 2 seasonal booster doses were tested for anti-circumsporozoite antibodies using enzyme-linked immunosorbent assay. The association between post-vaccination antibody titer and incidence of malaria was explored. RESULTS A strong anti-circumsporozoite antibody response to 3 priming doses of RTS,S/AS01E was seen (geometric mean titer, 368.9 enzyme-linked immunosorbent assay units/mL), but titers fell prior to the first booster dose. A strong antibody response to an annual, pre-malaria transmission season booster dose was observed, but this was lower than after the primary vaccination series and lower after the second than after the first booster dose (ratio of geometric mean rise, 0.66; 95% confidence interval [CI], .57-.77). Children whose antibody response was in the upper tercile post-vaccination had a lower incidence of malaria during the following year than children in the lowest tercile (hazard ratio, 0.43; 95% CI, .28-.66). CONCLUSIONS Seasonal vaccination with RTS,S/AS01E induced a strong booster antibody response that was lower after the second than after the first booster dose. The diminished antibody response to the second booster dose was not associated with diminished efficacy. CLINICAL TRIALS REGISTRATION NCT03143218.
Collapse
Affiliation(s)
| | | | - Matthew Cairns
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - Almahamoudou Mahamar
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Frédéric Nikièma
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Amadou Tapily
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | | | - Modibo Diarra
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Charles Zoungrana
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Djibrilla Issiaka
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Alassane Haro
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Koualy Sanogo
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Abdoul Aziz Sienou
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Mahamadou Kaya
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Seydou Traore
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Ismaila Thera
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Kalifa Diarra
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Amagana Dolo
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Irene Kuepfer
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Paul Snell
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Paul Milligan
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | | | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Abdoulaye Djimde
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | | | - Alassane Dicko
- The Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | | | - Brian Greenwood
- Correspondence: B. Greenwood, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel St., London WC1E 7HT, UK ()
| |
Collapse
|
12
|
Lipid Nanoparticle Delivery Systems to Enable mRNA-Based Therapeutics. Pharmaceutics 2022; 14:pharmaceutics14020398. [PMID: 35214130 PMCID: PMC8876479 DOI: 10.3390/pharmaceutics14020398] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/31/2022] [Accepted: 02/05/2022] [Indexed: 12/13/2022] Open
Abstract
The world raced to develop vaccines to protect against the rapid spread of SARS-CoV-2 infection upon the recognition of COVID-19 as a global pandemic. A broad spectrum of candidates was evaluated, with mRNA-based vaccines emerging as leaders due to how quickly they were available for emergency use while providing a high level of efficacy. As a modular technology, the mRNA-based vaccines benefitted from decades of advancements in both mRNA and delivery technology prior to the current global pandemic. The fundamental lessons of the utility of mRNA as a therapeutic were pioneered by Dr. Katalin Kariko and her colleagues, perhaps most notably in collaboration with Drew Weissman at University of Pennsylvania, and this foundational work paved the way for the development of the first ever mRNA-based therapeutic authorized for human use, COMIRNATY®. In this Special Issue of Pharmaceutics, we will be honoring Dr. Kariko for her great contributions to the mRNA technology to treat diseases with unmet needs. In this review article, we will focus on the delivery platform, the lipid nanoparticle (LNP) carrier, which allowed the potential of mRNA therapeutics to be realized. Similar to the mRNA technology, the development of LNP systems has been ongoing for decades before culminating in the success of the first clinically approved siRNA-LNP product, ONPATTRO®, a treatment for an otherwise fatal genetic disease called transthyretin amyloidosis. Lessons learned from the siRNA-LNP experience enabled the translation into the mRNA platform with the eventual authorization and approval of the mRNA-LNP vaccines against COVID-19. This marks the beginning of mRNA-LNP as a pharmaceutical option to treat genetic diseases.
Collapse
|
13
|
Wahl I, Wardemann H. How to induce protective humoral immunity against Plasmodium falciparum circumsporozoite protein. J Exp Med 2022; 219:212951. [PMID: 35006242 PMCID: PMC8754000 DOI: 10.1084/jem.20201313] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 12/03/2021] [Accepted: 12/17/2021] [Indexed: 12/23/2022] Open
Abstract
The induction of protective humoral immune responses against sporozoite surface proteins of the human parasite Plasmodium falciparum (Pf) is a prime goal in the development of a preerythrocytic malaria vaccine. The most promising antibody target is circumsporozoite protein (CSP). Although PfCSP induces strong humoral immune responses upon vaccination, vaccine efficacy is overall limited and not durable. Here, we review recent efforts to gain a better molecular and cellular understanding of anti-PfCSP B cell responses in humans and discuss ways to overcome limitations in the induction of stable titers of high-affinity antibodies that might help to increase vaccine efficacy and promote long-lived protection.
Collapse
Affiliation(s)
- Ilka Wahl
- B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Hedda Wardemann
- B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
14
|
Moncunill G, Carnes J, Chad Young W, Carpp L, De Rosa S, Campo JJ, Nhabomba A, Mpina M, Jairoce C, Finak G, Haas P, Muriel C, Van P, Sanz H, Dutta S, Mordmüller B, Agnandji ST, Díez-Padrisa N, Williams NA, Aponte JJ, Valim C, Neafsey DE, Daubenberger C, McElrath MJ, Dobaño C, Stuart K, Gottardo R. Transcriptional correlates of malaria in RTS,S/AS01-vaccinated African children: a matched case–control study. eLife 2022; 11:70393. [PMID: 35060479 PMCID: PMC8782572 DOI: 10.7554/elife.70393] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background: In a phase 3 trial in African infants and children, the RTS,S/AS01 vaccine (GSK) showed moderate efficacy against clinical malaria. We sought to further understand RTS,S/AS01-induced immune responses associated with vaccine protection. Methods: Applying the blood transcriptional module (BTM) framework, we characterized the transcriptomic response to RTS,S/AS01 vaccination in antigen-stimulated (and vehicle control) peripheral blood mononuclear cells sampled from a subset of trial participants at baseline and month 3 (1-month post-third dose). Using a matched case–control study design, we evaluated which of these ‘RTS,S/AS01 signature BTMs’ associated with malaria case status in RTS,S/AS01 vaccinees. Antigen-specific T-cell responses were analyzed by flow cytometry. We also performed a cross-study correlates analysis where we assessed the generalizability of our findings across three controlled human malaria infection studies of healthy, malaria-naive adult RTS,S/AS01 recipients. Results: RTS,S/AS01 vaccination was associated with downregulation of B-cell and monocyte-related BTMs and upregulation of T-cell-related BTMs, as well as higher month 3 (vs. baseline) circumsporozoite protein-specific CD4+ T-cell responses. There were few RTS,S/AS01-associated BTMs whose month 3 levels correlated with malaria risk. In contrast, baseline levels of BTMs associated with dendritic cells and with monocytes (among others) correlated with malaria risk. The baseline dendritic cell- and monocyte-related BTM correlations with malaria risk appeared to generalize to healthy, malaria-naive adults. Conclusions: A prevaccination transcriptomic signature associates with malaria in RTS,S/AS01-vaccinated African children, and elements of this signature may be broadly generalizable. The consistent presence of monocyte-related modules suggests that certain monocyte subsets may inhibit protective RTS,S/AS01-induced responses. Funding: Funding was obtained from the NIH-NIAID (R01AI095789), NIH-NIAID (U19AI128914), PATH Malaria Vaccine Initiative (MVI), and Ministerio de Economía y Competitividad (Instituto de Salud Carlos III, PI11/00423 and PI14/01422). The RNA-seq project has been funded in whole or in part with Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, under grant number U19AI110818 to the Broad Institute. This study was also supported by the Vaccine Statistical Support (Bill and Melinda Gates Foundation award INV-008576/OPP1154739 to R.G.). C.D. was the recipient of a Ramon y Cajal Contract from the Ministerio de Economía y Competitividad (RYC-2008-02631). G.M. was the recipient of a Sara Borrell–ISCIII fellowship (CD010/00156) and work was performed with the support of Department of Health, Catalan Government grant (SLT006/17/00109). This research is part of the ISGlobal’s Program on the Molecular Mechanisms of Malaria which is partially supported by the Fundación Ramón Areces and we acknowledge support from the Spanish Ministry of Science and Innovation through the ‘Centro de Excelencia Severo Ochoa 2019–2023’ Program (CEX2018-000806-S), and support from the Generalitat de Catalunya through the CERCA Program.
Collapse
Affiliation(s)
- Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona
- CIBER de Enfermedades Infecciosas
| | - Jason Carnes
- Center for Global Infectious Disease Research, Seattle Children's Research Institute
| | - William Chad Young
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Lindsay Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Stephen De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | | | - Augusto Nhabomba
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça
| | | | - Chenjerai Jairoce
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça
| | - Greg Finak
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Paige Haas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute
| | - Carl Muriel
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Phu Van
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Héctor Sanz
- ISGlobal, Hospital Clínic - Universitat de Barcelona
| | | | - Benjamin Mordmüller
- CIBER de Enfermedades Infecciosas
- Institute of Tropical Medicine and German Center for Infection Research
| | - Selidji T Agnandji
- Institute of Tropical Medicine and German Center for Infection Research
- Centre de Recherches Médicales de Lambaréné (CERMEL), BP 242
| | | | | | - John J Aponte
- ISGlobal, Hospital Clínic - Universitat de Barcelona
| | - Clarissa Valim
- Department of Global Health, Boston University School of Public Health
| | - Daniel E Neafsey
- Broad Institute of Massachusetts Institute of Technology and Harvard
- Harvard T.H. Chan School of Public Health
| | | | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- Departments of Laboratory Medicine and Medicine, University of Washington
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona
- CIBER de Enfermedades Infecciosas
| | - Ken Stuart
- Center for Global Infectious Disease Research, Seattle Children's Research Institute
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- Department of Pediatrics, University of Washington
- Department of Global Health, University of Washington
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- University of Lausanne and Centre Hospitalier Universitaire Vaudois
| |
Collapse
|
15
|
Seaton KE, Spreng RL, Abraha M, Reichartz M, Rojas M, Feely F, Huntwork RHC, Dutta S, Mudrak SV, Alam SM, Gregory S, Jongert E, Coccia M, Ulloa-Montoya F, Wille-Reece U, Tomaras GD, Dennison SM. Subclass and avidity of circumsporozoite protein specific antibodies associate with protection status against malaria infection. NPJ Vaccines 2021; 6:110. [PMID: 34462438 PMCID: PMC8405700 DOI: 10.1038/s41541-021-00372-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 07/30/2021] [Indexed: 12/19/2022] Open
Abstract
RTS,S/AS01 is an advanced pre-erythrocytic malaria vaccine candidate with demonstrated vaccine efficacy up to 86.7% in controlled human malaria infection (CHMI) studies; however, reproducible immune correlates of protection (CoP) are elusive. To identify candidates of humoral correlates of vaccine mediated protection, we measured antibody magnitude, subclass, and avidity for Plasmodium falciparum (Pf) circumsporozoite protein (CSP) by multiplex assays in two CHMI studies with varying RTS,S/AS01B vaccine dose and timing regimens. Central repeat (NANP6) IgG1 magnitude correlated best with protection status in univariate analyses and was the most predictive for protection in a multivariate model. NANP6 IgG3 magnitude, CSP IgG1 magnitude, and total serum antibody dissociation phase area-under-the-curve for NANP6, CSP, NPNA3, and N-interface binding were also associated with protection status in the regimen adjusted univariate analysis. Identification of multiple immune response features that associate with protection status, such as antibody subclasses, fine specificity and avidity reported here may accelerate development of highly efficacious vaccines against P. falciparum.
Collapse
Grants
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- United States Department of Defense | United States Army | Army Medical Command | Walter Reed Army Institute of Research (WRAIR)
- PATH Malaria Vaccine Initiative
- GlaxoSmithKline (GlaxoSmithKline plc.)
Collapse
Affiliation(s)
- Kelly E Seaton
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
- Duke University Department of Surgery, Durham, NC, USA.
| | - Rachel L Spreng
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
| | - Milite Abraha
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - Matthew Reichartz
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | | | - Frederick Feely
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - Richard H C Huntwork
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - Sheetij Dutta
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sarah V Mudrak
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke University Department of Pathology, Durham, NC, USA
| | - Scott Gregory
- PATH's Malaria Vaccine Initiative, Washington, DC, USA
| | | | | | | | - Ulrike Wille-Reece
- PATH's Malaria Vaccine Initiative, Washington, DC, USA
- GSK, Rockville, MD, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
- Duke University Department of Surgery, Durham, NC, USA.
- Duke University Department of Immunology, Durham, NC, USA.
- Duke University Department of Molecular Genetics and Microbiology, Durham, NC, USA.
| | - S Moses Dennison
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
- Duke University Department of Surgery, Durham, NC, USA.
| |
Collapse
|