1
|
Mitchell RA, Macià D, Jairoce C, Mpina M, Naidu A, Chopo-Pizarro A, Vázquez-Santiago M, Campo JJ, Aide P, Aguilar R, Daubenberger C, Dobaño C, Moncunill G. Effect of RTS,S/AS01 E vaccine booster dose on cellular immune responses in African infants and children. NPJ Vaccines 2024; 9:200. [PMID: 39455625 PMCID: PMC11511852 DOI: 10.1038/s41541-024-00977-y] [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: 05/08/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
RTS,S/AS01E, the first approved malaria vaccine, demonstrated moderate efficacy during the phase 3 pediatric trial. We previously investigated cell-mediated immune (CMI) responses following the primary 3-dose immunization and now report responses to the booster dose given 18 months later. Thirty CMI markers were measured by Luminex in supernatants of peripheral blood mononuclear cells from 709 children and infants after RTS,S/AS01E antigen stimulation, and their associations with malaria risk and antibodies one month post-booster and one year later were assessed. IL-2, IFN-γ, IL-17, IL-5, and IL-13 were associated with RTS,S/AS01E booster vaccination, and IL-2 responses to the circumsporozoite protein (CSP) remained higher after one year. IL-2 was associated with reduced malaria risk in one site, and IL-10 was associated with increased risk in infants. Anti-CSP IgG and IL-2 were moderately correlated one year after booster. This study highlights the moderate cell-mediated immunogenicity of the RTS,S/AS01E booster dose that aligns with partial recovery of RTS,S/AS01E vaccine efficacy.
Collapse
Affiliation(s)
- Robert A Mitchell
- ISGlobal, Barcelona, Catalonia, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Dídac Macià
- ISGlobal, Barcelona, Catalonia, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Chenjerai Jairoce
- ISGlobal, Barcelona, Catalonia, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Maxmillian Mpina
- Ifakara Health Institute. Bagamoyo Research and Training Centre, Bagamoyo, Tanzania
| | | | | | | | | | - Pedro Aide
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Ruth Aguilar
- ISGlobal, Barcelona, Catalonia, Spain
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain
| | - Claudia Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Carlota Dobaño
- ISGlobal, Barcelona, Catalonia, Spain.
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain.
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain.
| | - Gemma Moncunill
- ISGlobal, Barcelona, Catalonia, Spain.
- Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), Barcelona, Spain.
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain.
| |
Collapse
|
2
|
Friedman-Klabanoff DJ, Berry AA, Travassos MA, Shriver M, Cox C, Butts J, Lundeen JS, Strauss KA, Joshi S, Shrestha B, Mo AX, Nomicos EYH, Deye GA, Regules JA, Bergmann-Leitner ES, Pasetti MF, Laurens MB. Recombinant Full-length Plasmodium falciparum Circumsporozoite Protein-Based Vaccine Adjuvanted With Glucopyranosyl Lipid A-Liposome Quillaja saponaria 21: Results of Phase 1 Testing With Malaria Challenge. J Infect Dis 2024; 229:1883-1893. [PMID: 38330357 PMCID: PMC11175675 DOI: 10.1093/infdis/jiae062] [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: 08/26/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Malaria is preventable yet causes >600 000 deaths annually. RTS,S, the first marketed malaria vaccine, has modest efficacy, but improvements are needed for eradication. METHODS We conducted an open-label, dose escalation phase 1 study of a full-length recombinant circumsporozoite protein vaccine (rCSP) administered with adjuvant glucopyranosyl lipid A-liposome Quillaja saponaria 21 formulation (GLA-LSQ) on days 1, 29, and 85 or 1 and 490 to healthy, malaria-naive adults. The primary end points were safety and reactogenicity. The secondary end points were antibody responses and Plasmodium falciparum parasitemia after homologous controlled human malaria infection. RESULTS Participants were enrolled into 4 groups receiving rCSP/GLA-LSQ: 10 µg × 3 (n = 20), 30 µg × 3 (n = 10), 60 µg × 3 (n = 10), or 60 µg × 2 (n = 9); 10 participants received 30 µg rCSP alone × 3, and there were 6 infectivity controls. Participants experienced no serious adverse events. Rates of solicited and unsolicited adverse events were similar among groups. All 26 participants who underwent controlled human malaria infection 28 days after final vaccinations developed malaria. Increasing vaccine doses induced higher immunoglobulin G titers but did not achieve previously established RTS,S benchmarks. CONCLUSIONS rCSP/GLA-LSQ had favorable safety results. However, tested regimens did not induce protective immunity. Further investigation could assess whether adjuvant or schedule adjustments improve efficacy. CLINICAL TRIALS REGISTRATION NCT03589794.
Collapse
Affiliation(s)
- DeAnna J Friedman-Klabanoff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Andrea A Berry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mark A Travassos
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mallory Shriver
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | - Kathleen A Strauss
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sudhaunshu Joshi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Biraj Shrestha
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Annie X Mo
- Parasitology and International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Effie Y H Nomicos
- Parasitology and International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Gregory A Deye
- Parasitology and International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jason A Regules
- Biologics Research & Development, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Elke S Bergmann-Leitner
- Biologics Research & Development, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Marcela F Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Matthew B Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Nebie I, Palacpac NMQ, Bougouma EC, Diarra A, Ouédraogo A, D’Alessio F, Houard S, Tiono AB, Cousens S, Horii T, Sirima SB. Persistence of Anti-SE36 Antibodies Induced by the Malaria Vaccine Candidate BK-SE36/CpG in 5-10-Year-Old Burkinabe Children Naturally Exposed to Malaria. Vaccines (Basel) 2024; 12:166. [PMID: 38400149 PMCID: PMC10892924 DOI: 10.3390/vaccines12020166] [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: 12/15/2023] [Revised: 01/25/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
Information on the dynamics and decline/persistence of antibody titres is important in vaccine development. A recent vaccine trial in malaria-exposed, healthy African adults and children living in a malaria hyperendemic and seasonal area (Ouagadougou, Burkina Faso) was the first study in which BK-SE36/CpG was administered to different age groups. In 5- to 10-year-old children, the risk of malaria infection was markedly lower in the BK-SE36/CpG arm compared to the control arm. We report here data on antibody titres measured in this age-group after the high malaria transmission season of 2021 (three years after the first vaccine dose was administered). At Year 3, 83% of children had detectable anti-SE36 total IgG antibodies. Geometric mean antibody titres and the proportion of children with detectable anti-SE36 antibodies were markedly higher in the BK-SE36/CpG arm than the control (rabies) arm. The information obtained in this study will guide investigators on future vaccine/booster schedules for this promising blood-stage malaria vaccine candidate.
Collapse
Affiliation(s)
- Issa Nebie
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 10248, Burkina Faso; (I.N.); (E.C.B.); (A.D.); (A.O.); (A.B.T.)
| | - Nirianne Marie Q. Palacpac
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Osaka, Japan;
| | - Edith Christiane Bougouma
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 10248, Burkina Faso; (I.N.); (E.C.B.); (A.D.); (A.O.); (A.B.T.)
| | - Amidou Diarra
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 10248, Burkina Faso; (I.N.); (E.C.B.); (A.D.); (A.O.); (A.B.T.)
| | - Alphonse Ouédraogo
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 10248, Burkina Faso; (I.N.); (E.C.B.); (A.D.); (A.O.); (A.B.T.)
| | - Flavia D’Alessio
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Voßstraße 2, 69115 Heidelberg, Germany; (F.D.); (S.H.)
| | - Sophie Houard
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Voßstraße 2, 69115 Heidelberg, Germany; (F.D.); (S.H.)
| | - Alfred B. Tiono
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 10248, Burkina Faso; (I.N.); (E.C.B.); (A.D.); (A.O.); (A.B.T.)
| | - Simon Cousens
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK;
| | - Toshihiro Horii
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Osaka, Japan;
| | - Sodiomon B. Sirima
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 10248, Burkina Faso; (I.N.); (E.C.B.); (A.D.); (A.O.); (A.B.T.)
| |
Collapse
|
4
|
Martin GM, Torres JL, Pholcharee T, Oyen D, Flores-Garcia Y, Gibson G, Moskovitz R, Beutler N, Jung DD, Copps J, Lee WH, Gonzalez-Paez G, Emerling D, MacGill RS, Locke E, King CR, Zavala F, Wilson IA, Ward AB. Affinity-matured homotypic interactions induce spectrum of PfCSP structures that influence protection from malaria infection. Nat Commun 2023; 14:4546. [PMID: 37507365 PMCID: PMC10382551 DOI: 10.1038/s41467-023-40151-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The generation of high-quality antibody responses to Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP), the primary surface antigen of Pf sporozoites, is paramount to the development of an effective malaria vaccine. Here we present an in-depth structural and functional analysis of a panel of potent antibodies encoded by the immunoglobulin heavy chain variable (IGHV) gene IGHV3-33, which is among the most prevalent and potent antibody families induced in the anti-PfCSP immune response and targets the Asn-Ala-Asn-Pro (NANP) repeat region. Cryo-electron microscopy (cryo-EM) reveals a remarkable spectrum of helical antibody-PfCSP structures stabilized by homotypic interactions between tightly packed fragments antigen binding (Fabs), many of which correlate with somatic hypermutation. We demonstrate a key role of these mutated homotypic contacts for high avidity binding to PfCSP and in protection from Pf malaria infection. Together, these data emphasize the importance of anti-homotypic affinity maturation in the frequent selection of IGHV3-33 antibodies and highlight key features underlying the potent protection of this antibody family.
Collapse
Affiliation(s)
- Gregory M Martin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jonathan L Torres
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Tossapol Pholcharee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Biochemistry, University of Oxford, Oxford, OX1 3DR, UK
| | - David Oyen
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Pfizer Inc, San Diego, CA, 92121, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Grace Gibson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Re'em Moskovitz
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Diana D Jung
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Gonzalo Gonzalez-Paez
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | | | | | - Emily Locke
- PATH's Malaria Vaccine Initiative, Washington, DC, 20001, USA
| | - C Richter King
- PATH's Malaria Vaccine Initiative, Washington, DC, 20001, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
5
|
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: 4.0] [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
|
6
|
Ngulube P. Humoral Immune Responses to P. falciparum Circumsporozoite Protein (Pfcsp) Induced by the RTS, S Vaccine - Current Update. Infect Drug Resist 2023; 16:2147-2157. [PMID: 37077252 PMCID: PMC10106824 DOI: 10.2147/idr.s401247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/23/2023] [Indexed: 04/21/2023] Open
Abstract
Malaria vaccines targeting the circumsporozoite protein (CSP) of the P. falciparum parasite have been overall relatively promising. RTS, S is a pre-erythrocytic recombinant protein-based malaria vaccine that targets CSP. RTS, S effectiveness shows some limited success regardless of its 58% efficacy for severe disease. P. falciparum circumsporozoite protein (Pfcsp) has stood to be the main candidate protein for most pre-erythrocytic stage vaccines. Studies on the structural and biophysical characteristics of antibodies specific to CSP (anti-CSP) are underway to achieve fine specificity with the CSP polymorphic regions. More recent studies have proposed the use of different kinds of monoclonal antibodies, the use of appropriate adjuvants, ideal vaccination dose and frequency, and improved targeting of particular epitopes for the robust production of functional antibodies and high complement-fixing activity as other potential methods for achieving long-lasting RTS, S. This review highlights recent findings regarding humoral immune responses to CSP elicited by RTS, S vaccine.
Collapse
Affiliation(s)
- Peter Ngulube
- Department of Biological Sciences, Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
- Correspondence: Peter Ngulube, Email
| |
Collapse
|
7
|
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: 11] [Impact Index Per Article: 5.5] [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
|
8
|
Tursi NJ, Reeder SM, Flores-Garcia Y, Bah MA, Mathis-Torres S, Salgado-Jimenez B, Esquivel R, Xu Z, Chu JD, Humeau L, Patel A, Zavala F, Weiner DB. Engineered DNA-encoded monoclonal antibodies targeting Plasmodium falciparum circumsporozoite protein confer single dose protection in a murine malaria challenge model. Sci Rep 2022; 12:14313. [PMID: 35995959 PMCID: PMC9395511 DOI: 10.1038/s41598-022-18375-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/06/2022] [Indexed: 12/15/2022] Open
Abstract
Novel approaches for malaria prophylaxis remain important. Synthetic DNA-encoded monoclonal antibodies (DMAbs) are a promising approach to generate rapid, direct in vivo host-generated mAbs with potential benefits in production simplicity and distribution coupled with genetic engineering. Here, we explore this approach in a malaria challenge model. We engineered germline-reverted DMAbs based on human mAb clones CIS43, 317, and L9 which target a junctional epitope, major repeat, and minor repeat of the Plasmodium falciparum circumsporozoite protein (CSP) respectively. DMAb variants were encoded into a plasmid vector backbone and their expression and binding profiles were characterized. We demonstrate long-term serological expression of DMAb constructs resulting in in vivo efficacy of CIS43 GL and 317 GL in a rigorous mosquito bite mouse challenge model. Additionally, we engineered an Fc modified variant of CIS43 and L9-based DMAbs to ablate binding to C1q to test the impact of complement-dependent Fc function on challenge outcomes. Complement knockout variant DMAbs demonstrated similar protection to that of WT Fc DMAbs supporting the notion that direct binding to the parasite is sufficient for the protection observed. Further investigation of DMAbs for malaria prophylaxis appears of importance.
Collapse
Affiliation(s)
- Nicholas J Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sophia M Reeder
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mamadou A Bah
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Shamika Mathis-Torres
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Berenice Salgado-Jimenez
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Rianne Esquivel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Ziyang Xu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jacqueline D Chu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Laurent Humeau
- Inovio Pharmaceuticals, Plymouth Meeting, PA, 19462, USA
| | - Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
9
|
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: 7] [Impact Index Per Article: 3.5] [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
|
10
|
Kandi V, Suvvari TK, Vadakedath S, Godishala V. Microbes, Clinical trials, Drug Discovery, and Vaccine Development: The Current Perspectives. BORNEO JOURNAL OF PHARMACY 2021. [DOI: 10.33084/bjop.v4i4.2571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Because of the frequent emergence of novel microbial species and the re-emergence of genetic variants of hitherto known microbes, the global healthcare system, and human health has been thrown into jeopardy. Also, certain microbes that possess the ability to develop multi-drug resistance (MDR) have limited the treatment options in cases of serious infections, and increased hospital and treatment costs, and associated morbidity and mortality. The recent discovery of the novel Coronavirus (n-CoV), the Severe Acute Respiratory Syndrome CoV-2 (SARS-CoV-2) that is causing the CoV Disease-19 (COVID-19) has resulted in severe morbidity and mortality throughout the world affecting normal human lives. The major concern with the current pandemic is the non-availability of specific drugs and an incomplete understanding of the pathobiology of the virus. It is therefore important for pharmaceutical establishments to envisage the discovery of therapeutic interventions and potential vaccines against the novel and MDR microbes. Therefore, this review is attempted to update and explore the current perspectives in microbes, clinical research, drug discovery, and vaccine development to effectively combat the emerging novel and re-emerging genetic variants of microbes.
Collapse
|
11
|
Systems analysis and controlled malaria infection in Europeans and Africans elucidate naturally acquired immunity. Nat Immunol 2021; 22:654-665. [PMID: 33888898 DOI: 10.1038/s41590-021-00911-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/02/2021] [Indexed: 01/31/2023]
Abstract
Controlled human infections provide opportunities to study the interaction between the immune system and malaria parasites, which is essential for vaccine development. Here, we compared immune signatures of malaria-naive Europeans and of Africans with lifelong malaria exposure using mass cytometry, RNA sequencing and data integration, before and 5 and 11 days after venous inoculation with Plasmodium falciparum sporozoites. We observed differences in immune cell populations, antigen-specific responses and gene expression profiles between Europeans and Africans and among Africans with differing degrees of immunity. Before inoculation, an activated/differentiated state of both innate and adaptive cells, including elevated CD161+CD4+ T cells and interferon-γ production, predicted Africans capable of controlling parasitemia. After inoculation, the rapidity of the transcriptional response and clusters of CD4+ T cells, plasmacytoid dendritic cells and innate T cells were among the features distinguishing Africans capable of controlling parasitemia from susceptible individuals. These findings can guide the development of a vaccine effective in malaria-endemic regions.
Collapse
|
12
|
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: 1.3] [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
|
13
|
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
|
14
|
Tomlinson A, Semblat JP, Gamain B, Chêne A. VAR2CSA-Mediated Host Defense Evasion of Plasmodium falciparum Infected Erythrocytes in Placental Malaria. Front Immunol 2021; 11:624126. [PMID: 33633743 PMCID: PMC7900151 DOI: 10.3389/fimmu.2020.624126] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/23/2020] [Indexed: 12/04/2022] Open
Abstract
Over 30 million women living in P. falciparum endemic areas are at risk of developing malaria during pregnancy every year. Placental malaria is characterized by massive accumulation of infected erythrocytes in the intervillous space of the placenta, accompanied by infiltration of immune cells, particularly monocytes. The consequent local inflammation and the obstruction of the maternofetal exchanges can lead to severe clinical outcomes for both mother and child. Even if protection against the disease can gradually be acquired following successive pregnancies, the malaria parasite has developed a large panel of evasion mechanisms to escape from host defense mechanisms and manipulate the immune system to its advantage. Infected erythrocytes isolated from placentas of women suffering from placental malaria present a unique phenotype and express the pregnancy-specific variant VAR2CSA of the Plasmodium falciparum Erythrocyte Membrane Protein (PfEMP1) family at their surface. The polymorphic VAR2CSA protein is able to mediate the interaction of infected erythrocytes with a variety of host cells including placental syncytiotrophoblasts and leukocytes but also with components of the immune system such as non-specific IgM. This review summarizes the described VAR2CSA-mediated host defense evasion mechanisms employed by the parasite during placental malaria to ensure its survival and persistence.
Collapse
Affiliation(s)
- Alice Tomlinson
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Jean-Philippe Semblat
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Benoît Gamain
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Arnaud Chêne
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France.,Laboratory of Excellence GR-Ex, Paris, France
| |
Collapse
|
15
|
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
|
16
|
Kucharska I, Thai E, Srivastava A, Rubinstein JL, Pomès R, Julien JP. Structural ordering of the Plasmodium berghei circumsporozoite protein repeats by inhibitory antibody 3D11. eLife 2020; 9:e59018. [PMID: 33253113 PMCID: PMC7704109 DOI: 10.7554/elife.59018] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 11/04/2020] [Indexed: 12/15/2022] Open
Abstract
Plasmodium sporozoites express circumsporozoite protein (CSP) on their surface, an essential protein that contains central repeating motifs. Antibodies targeting this region can neutralize infection, and the partial efficacy of RTS,S/AS01 - the leading malaria vaccine against P. falciparum (Pf) - has been associated with the humoral response against the repeats. Although structural details of antibody recognition of PfCSP have recently emerged, the molecular basis of antibody-mediated inhibition of other Plasmodium species via CSP binding remains unclear. Here, we analyze the structure and molecular interactions of potent monoclonal antibody (mAb) 3D11 binding to P. berghei CSP (PbCSP) using molecular dynamics simulations, X-ray crystallography, and cryoEM. We reveal that mAb 3D11 can accommodate all subtle variances of the PbCSP repeating motifs, and, upon binding, induces structural ordering of PbCSP through homotypic interactions. Together, our findings uncover common mechanisms of antibody evolution in mammals against the CSP repeats of Plasmodium sporozoites.
Collapse
Affiliation(s)
- Iga Kucharska
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
| | - Elaine Thai
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
| | - Ananya Srivastava
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
| | - John L Rubinstein
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
- Department of Medical Biophysics, University of TorontoTorontoCanada
| | - Régis Pomès
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
- Department of Immunology, University of TorontoTorontoCanada
| |
Collapse
|
17
|
Mordmüller B, Sulyok M, Egger-Adam D, Resende M, de Jongh WA, Jensen MH, Smedegaard HH, Ditlev SB, Soegaard M, Poulsen L, Dyring C, Calle CL, Knoblich A, Ibáñez J, Esen M, Deloron P, Ndam N, Issifou S, Houard S, Howard RF, Reed SG, Leroy O, Luty AJF, Theander TG, Kremsner PG, Salanti A, Nielsen MA. First-in-human, Randomized, Double-blind Clinical Trial of Differentially Adjuvanted PAMVAC, A Vaccine Candidate to Prevent Pregnancy-associated Malaria. Clin Infect Dis 2020; 69:1509-1516. [PMID: 30629148 PMCID: PMC6792113 DOI: 10.1093/cid/ciy1140] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 01/03/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Malaria in pregnancy has major impacts on mother and child health. To complement existing interventions, such as intermittent preventive treatment and use of impregnated bed nets, we developed a malaria vaccine candidate with the aim of reducing sequestration of asexual "blood-stage" parasites in the placenta, the major virulence mechanism. METHODS The vaccine candidate PAMVAC is based on a recombinant fragment of VAR2CSA, the Plasmodium falciparum protein responsible for binding to the placenta via chondroitin sulfate A (CSA). Healthy, adult malaria-naive volunteers were immunized with 3 intramuscular injections of 20 μg (n = 9) or 50 μg (n = 27) PAMVAC, adjuvanted with Alhydrogel or glucopyranosyl lipid adjuvant in stable emulsion (GLA-SE) or in a liposomal formulation with QS21 (GLA-LSQ). Allocation was random and double blind. The vaccine was given every 4 weeks. Volunteers were observed for 6 months following last immunization. RESULTS All PAMVAC formulations were safe and well tolerated. A total of 262 adverse events (AEs) occurred, 94 (10 grade 2 and 2 grade 3) at least possibly related to the vaccine. No serious AEs occurred. Distribution and severity of AEs were similar in all arms. PAMVAC was immunogenic in all participants. PAMVAC-specific antibody levels were highest with PAMVAC-GLA-SE. The antibodies inhibited binding of VAR2CSA expressing P. falciparum-infected erythrocytes to CSA in a standardized functional assay. CONCLUSIONS PAMVAC formulated with Alhydrogel or GLA-based adjuvants was safe, well tolerated, and induced functionally active antibodies. Next, PAMVAC will be assessed in women before first pregnancies in an endemic area. CLINICAL TRIALS REGISTRATION EudraCT 2015-001827-21; ClinicalTrials.gov NCT02647489.
Collapse
Affiliation(s)
- Benjamin Mordmüller
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany.,Centre de Recherches Médicales de Lambaréné, Gabon
| | - Mihály Sulyok
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany
| | - Diane Egger-Adam
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany
| | - Mafalda Resende
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | | | - Mette H Jensen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | - Helle Holm Smedegaard
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | - Sisse B Ditlev
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | | | | | | | - Carlos Lamsfus Calle
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany
| | - Annette Knoblich
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany
| | - Javier Ibáñez
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany
| | - Meral Esen
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany.,Centre de Recherches Médicales de Lambaréné, Gabon
| | - Philippe Deloron
- Mère et Enfant face aux Infections Tropicales, Institut de Recherche pour le Développement, Université Paris 5, Sorbonne Paris Cité, France
| | - Nicaise Ndam
- Mère et Enfant face aux Infections Tropicales, Institut de Recherche pour le Développement, Université Paris 5, Sorbonne Paris Cité, France
| | - Saadou Issifou
- Fondation pour la Recherche Scientifique and Institut de Recherche Clinique du Bénin, Cotonou
| | | | | | - Steven G Reed
- Infectious Disease Research Institute, Seattle, Washington
| | - Odile Leroy
- European Vaccine Initiative, Heidelberg, Germany
| | - Adrian J F Luty
- Mère et Enfant face aux Infections Tropicales, Institut de Recherche pour le Développement, Université Paris 5, Sorbonne Paris Cité, France
| | - Thor G Theander
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | - Peter G Kremsner
- Institut für Tropenmedizin, Universitätsklinikum Tübingen and Deutsches Zentrum für Infektionsforschung, Germany.,Centre de Recherches Médicales de Lambaréné, Gabon
| | - Ali Salanti
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| | - Morten A Nielsen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital
| |
Collapse
|
18
|
Skwarczynski M, Chandrudu S, Rigau-Planella B, Islam MT, Cheong YS, Liu G, Wang X, Toth I, Hussein WM. Progress in the Development of Subunit Vaccines against Malaria. Vaccines (Basel) 2020; 8:vaccines8030373. [PMID: 32664421 PMCID: PMC7563759 DOI: 10.3390/vaccines8030373] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/02/2022] Open
Abstract
Malaria is a life-threatening disease and one of the main causes of morbidity and mortality in the human population. The disease also results in a major socio-economic burden. The rapid spread of malaria epidemics in developing countries is exacerbated by the rise in drug-resistant parasites and insecticide-resistant mosquitoes. At present, malaria research is focused mainly on the development of drugs with increased therapeutic effects against Plasmodium parasites. However, a vaccine against the disease is preferable over treatment to achieve long-term control. Trials to develop a safe and effective immunization protocol for the control of malaria have been occurring for decades, and continue on today; still, no effective vaccines are available on the market. Recently, peptide-based vaccines have become an attractive alternative approach. These vaccines utilize short protein fragments to induce immune responses against malaria parasites. Peptide-based vaccines are safer than traditional vaccines, relatively inexpensive to produce, and can be composed of multiple T- and B-cell epitopes integrated into one antigenic formulation. Various combinations, based on antigen choice, peptide epitope modification and delivery mechanism, have resulted in numerous potential malaria vaccines candidates; these are presently being studied in both preclinical and clinical trials. This review describes the current landscape of peptide-based vaccines, and addresses obstacles and opportunities in the production of malaria vaccines.
Collapse
Affiliation(s)
- Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Saranya Chandrudu
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Berta Rigau-Planella
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Md. Tanjir Islam
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Yee S. Cheong
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Genan Liu
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
| | - Xiumin Wang
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Correspondence: (I.T.); (W.M.H.)
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (M.S.); (S.C.); (B.R.-P.); (M.T.I.); (Y.S.C.); (G.L.); (X.W.)
- Correspondence: (I.T.); (W.M.H.)
| |
Collapse
|
19
|
Mitran CJ, Yanow SK. The Case for Exploiting Cross-Species Epitopes in Malaria Vaccine Design. Front Immunol 2020; 11:335. [PMID: 32174924 PMCID: PMC7056716 DOI: 10.3389/fimmu.2020.00335] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/10/2020] [Indexed: 12/21/2022] Open
Abstract
The infection dynamics between different species of Plasmodium that infect the same human host can both suppress and exacerbate disease. This could arise from inter-parasite interactions, such as competition, from immune regulation, or both. The occurrence of protective, cross-species (heterologous) immunity is an unlikely event, especially considering that strain-transcending immunity within a species is only partial despite lifelong exposure to that species. Here we review the literature in humans and animal models to identify the contexts where heterologous immunity can arise, and which antigens may be involved. From the perspective of vaccine design, understanding the mechanisms by which exposure to an antigen from one species can elicit a protective response to another species offers an alternative strategy to conventional approaches that focus on immunodominant antigens within a single species. The underlying hypothesis is that certain epitopes are conserved across evolution, in sequence or in structure, and shared in antigens from different species. Vaccines that focus on conserved epitopes may overcome the challenges posed by polymorphic immunodominant antigens; but to uncover these epitopes requires approaches that consider the evolutionary history of protein families across species. The key question for vaccinologists will be whether vaccines that express these epitopes can elicit immune responses that are functional and contribute to protection against Plasmodium parasites.
Collapse
Affiliation(s)
| | - Stephanie K. Yanow
- School of Public Health, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
20
|
Plotkin SA. Updates on immunologic correlates of vaccine-induced protection. Vaccine 2019; 38:2250-2257. [PMID: 31767462 DOI: 10.1016/j.vaccine.2019.10.046] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023]
Abstract
Correlates of protection (CoPs) are increasingly important in the development and licensure of vaccines. Although the study of CoPs was initially directed at identifying a single immune function that could explain vaccine efficacy, it has become increasingly clear that there are often multiple functions responsible for efficacy. This review is meant to supplement prior articles on the subject, illustrating both simple and complex CoPs.
Collapse
Affiliation(s)
- Stanley A Plotkin
- Emeritus Professor of Pediatrics, University of Pennsylvania, Vaxconsult, 4650 Wismer Rd., Doylestown, PA 18902, United States.
| |
Collapse
|
21
|
Julien JP, Wardemann H. Antibodies against Plasmodium falciparum malaria at the molecular level. Nat Rev Immunol 2019; 19:761-775. [DOI: 10.1038/s41577-019-0209-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
|
22
|
Ottenhoff THM. Correlates of vaccine adjuvanticity, vaccine activity, protective immunity and disease in human infectious disease and cancer. Semin Immunol 2018; 39:1-3. [PMID: 30318307 DOI: 10.1016/j.smim.2018.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Tom H M Ottenhoff
- Dept. Infectious Diseases, Leiden University Medical Center, P.O. Box 9600, 2300RC, Leiden, the Netherlands.
| |
Collapse
|