1
|
Tajudeen YA, Oladipo HJ, Yusuff SI, Abimbola SO, Abdulkadir M, Oladunjoye IO, Omotosho AO, Egbewande OM, Shittu HD, Yusuf RO, Ogundipe O, Muili AO, Afolabi AO, Dahesh SMA, Gameil MAM, El-Sherbini MS. A landscape review of malaria vaccine candidates in the pipeline. Trop Dis Travel Med Vaccines 2024; 10:19. [PMID: 39085983 PMCID: PMC11293096 DOI: 10.1186/s40794-024-00222-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 04/15/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Globally, malaria continues to pose a major health challenge, with approximately 247 million cases of the illness and 627,000 deaths reported in 2021. However, the threat is particularly pronounced in sub-Saharan African countries, where pregnant women and children under the age of five face heightened vulnerability to the disease. As a result, the imperative to develop malaria vaccines especially for these vulnerable populations, remains crucial in the pursuit of malaria eradication. However, despite decades of research, effective vaccine development faces technical challenges, including the rapid spread of drug-resistant parasite strains, the complex parasite lifecycle, the development of liver hypnozoites with potential for relapse, and evasion of the host immune system. This review aims to discuss the different malaria vaccine candidates in the pipeline, highlighting different approaches used for adjuvating these candidates, their benefits, and outcomes, and summarizing the progress of these vaccine candidates under development. METHOD A comprehensive web-based search for peer-reviewed journal articles published in SCOPUS, MEDLINE (via PubMed), Science Direct, WHO, and Advanced Google Scholar databases was conducted from 1990 to May 2022. Context-specific keywords such as "Malaria", "Malaria Vaccine", "Malaria Vaccine Candidates", "Vaccine Development", "Vaccine Safety", "Clinical Trials", "mRNA Vaccines", "Viral Vector Vaccines", "Protein-based Vaccines", "Subunit Vaccines", "Vaccine Adjuvants", "Vaccine-induced Immune Responses", and "Immunogenicity" were emphatically considered. Articles not directly related to malaria vaccine candidates in preclinical and clinical stages of development were excluded. RESULTS Various approaches have been studied for malaria vaccine development, targeting different parasite lifecycle stages, including the pre-erythrocytic, erythrocytic, and sexual stages. The RTS, S/AS01 vaccine, the first human parasite vaccine reaching WHO-listed authority maturity level 4, has demonstrated efficacy in preventing clinical malaria in African children. However, progress was slow in introducing other safe, and feasible malaria vaccines through clinical trials . Recent studies highlight the potential effectiveness of combining pre-erythrocytic and blood-stage vaccines, along with the advantages of mRNA vaccines for prophylaxis and treatment, and nonstructural vaccines for large-scale production. CONCLUSION Malaria vaccine candidates targeting different lifecycle stages of the parasite range from chemoprophylaxis vaccination to cross-species immune protection. The use of a multi-antigen, multi-stage combinational vaccine is therefore essential in the context of global health. This demands careful understanding and critical consideration of the long-term multi-faceted interplay of immune interference, co-dominance, complementary immune response, molecular targets, and adjuvants affecting the overall vaccine-induced immune response. Despite challenges, advancements in clinical trials and vaccination technology offer promising possibilities for novel approaches in malaria vaccine development.
Collapse
Affiliation(s)
- Yusuf Amuda Tajudeen
- Department of Microbiology, Faculty of Life Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
- Department of Epidemiology and Medical Statistics, Faculty of Public Health, College of Medicine, University of Ibadan, P.M.B 5017 G.P.O, Ibadan, Oyo State, Nigeria
| | - Habeebullah Jayeola Oladipo
- Department of Microbiology, Faculty of Life Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
- Faculty of Pharmaceutical Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
| | - Sodiq Inaolaji Yusuff
- Department of Medicine, Faculty of Clinical Sciences, Obafemi Awolowo University, Ibadan- Ife Rd, Ife, 220282, Osun State, Nigeria
| | - Samuel O Abimbola
- Cyprus International Institute of Environmental and Public Health, Cyprus University of Technology, Limassol, 3036, Cyprus
| | - Muritala Abdulkadir
- Faculty of Pharmaceutical Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
| | - Iyiola Olatunji Oladunjoye
- Department of Microbiology, Faculty of Life Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
| | - Abass Olawale Omotosho
- Department of Microbiology, Faculty of Pure and Applied Sciences, Kwara State University, P.M.B 1530, Malete-Ilorin, Ilorin, Nigeria
| | | | | | - Rashidat Onyinoyi Yusuf
- Faculty of Pharmaceutical Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
| | - Oluwatosin Ogundipe
- Department of Epidemiology and Medical Statistics, Faculty of Public Health, College of Medicine, University of Ibadan, P.M.B 5017 G.P.O, Ibadan, Oyo State, Nigeria
| | - Abdulbasit Opeyemi Muili
- Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, P.M.B 4000, Ogbomosho, Oyo State, Nigeria
| | - Abdullateef Opeyemi Afolabi
- Faculty of Biomedical Sciences, Department of Microbiology and Immunology, Kampala International University, Bushenyi, Uganda.
| | - Salwa M A Dahesh
- Research Institute of Medical Entomology, General Organization for Teaching Hospitals and Institutes, GOTHI, Damietta, Egypt
| | | | - Mona Said El-Sherbini
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
2
|
Tottey S, Shoji Y, Mark Jones R, Musiychuk K, Chichester JA, Miura K, Zhou L, Lee SM, Plieskatt J, Wu Y, Long CA, Streatfield SJ, Yusibov V. Engineering of a plant-produced virus-like particle to improve the display of the Plasmodium falciparum Pfs25 antigen and transmission-blocking activity of the vaccine candidate. Vaccine 2023; 41:938-944. [PMID: 36585278 PMCID: PMC9888754 DOI: 10.1016/j.vaccine.2022.12.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 12/04/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Malaria kills around 409,000 people a year, mostly children under the age of five. Malaria transmission-blocking vaccines work to reduce malaria prevalence in a community and have the potential to be part of a multifaceted approach required to eliminate the parasites causing the disease. Pfs25 is a leading malaria transmission-blocking antigen and has been successfully produced in a plant expression system as both a subunit vaccine and as a virus-like particle. This study demonstrates an improved version of the virus-like particle antigen display molecule by eliminating known protease sites from the prior A85 variant. This re-engineered molecule, termed B29, displays three times the number of Pfs25 antigens per virus-like particle compared to the original Pfs25 virus-like particle. An improved purification scheme was also developed, resulting in a substantially higher yield and improved purity. The molecule was evaluated in a mouse model and found to induce improved transmission-blocking activity at lower doses and longer durations than the original molecule.
Collapse
Affiliation(s)
- Stephen Tottey
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| | - Yoko Shoji
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| | - R Mark Jones
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| | - Konstantin Musiychuk
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| | - Jessica A Chichester
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Luwen Zhou
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Shwu-Maan Lee
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | | | - Yimin Wu
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Stephen J Streatfield
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA.
| | - Vidadi Yusibov
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| |
Collapse
|
3
|
Reuling IJ, Mendes AM, de Jong GM, Fabra-García A, Nunes-Cabaço H, van Gemert GJ, Graumans W, Coffeng LE, de Vlas SJ, Yang ASP, Lee C, Wu Y, Birkett AJ, Ockenhouse CF, Koelewijn R, van Hellemond JJ, van Genderen PJJ, Sauerwein RW, Prudêncio M. An open-label phase 1/2a trial of a genetically modified rodent malaria parasite for immunization against Plasmodium falciparum malaria. Sci Transl Med 2021; 12:12/544/eaay2578. [PMID: 32434846 DOI: 10.1126/scitranslmed.aay2578] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
For some diseases, successful vaccines have been developed using a nonpathogenic counterpart of the causative microorganism of choice. The nonpathogenicity of the rodent Plasmodium berghei (Pb) parasite in humans prompted us to evaluate its potential as a platform for vaccination against human infection by Plasmodium falciparum (Pf), a causative agent of malaria. We hypothesized that the genetic insertion of a leading protein target for clinical development of a malaria vaccine, Pf circumsporozoite protein (CSP), in its natural pre-erythrocytic environment, would enhance Pb's capacity to induce protective immunity against Pf infection. Hence, we recently generated a transgenic Pb sporozoite immunization platform expressing PfCSP (PbVac), and we now report the clinical evaluation of its biological activity against controlled human malaria infection (CHMI). This first-in-human trial shows that PbVac is safe and well tolerated, when administered by a total of ~300 PbVac-infected mosquitoes per volunteer. Although protective efficacy evaluated by CHMI showed no sterile protection at the tested dose, significant delays in patency (2.2 days, P = 0.03) and decreased parasite density were observed after immunization, corresponding to an estimated 95% reduction in Pf liver parasite burden (confidence interval, 56 to 99%; P = 0.010). PbVac elicits dose-dependent cross-species cellular immune responses and functional PfCSP-dependent antibody responses that efficiently block Pf sporozoite invasion of liver cells in vitro. This study demonstrates that PbVac immunization elicits a marked biological effect, inhibiting a subsequent infection by the human Pf parasite, and establishes the clinical validation of a new paradigm in malaria vaccination.
Collapse
Affiliation(s)
- Isaie J Reuling
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - António M Mendes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Gerdie M de Jong
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 GD Rotterdam, Netherlands
| | - Amanda Fabra-García
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Helena Nunes-Cabaço
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Geert-Jan van Gemert
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Wouter Graumans
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Luc E Coffeng
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, Netherlands
| | - Sake J de Vlas
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, Netherlands
| | - Annie S P Yang
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Cynthia Lee
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | - Yimin Wu
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | | | | | - Rob Koelewijn
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 GD Rotterdam, Netherlands
| | - Jaap J van Hellemond
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 GD Rotterdam, Netherlands
| | - Perry J J van Genderen
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 3015 GD Rotterdam, Netherlands. .,Corporate Travel Clinic Erasmus MC, 3015 CP Rotterdam, Netherlands
| | - Robert W Sauerwein
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands.
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal.
| |
Collapse
|
4
|
Suscovich TJ, Fallon JK, Das J, Demas AR, Crain J, Linde CH, Michell A, Natarajan H, Arevalo C, Broge T, Linnekin T, Kulkarni V, Lu R, Slein MD, Luedemann C, Marquette M, March S, Weiner J, Gregory S, Coccia M, Flores-Garcia Y, Zavala F, Ackerman ME, Bergmann-Leitner E, Hendriks J, Sadoff J, Dutta S, Bhatia SN, Lauffenburger DA, Jongert E, Wille-Reece U, Alter G. Mapping functional humoral correlates of protection against malaria challenge following RTS,S/AS01 vaccination. Sci Transl Med 2021; 12:12/553/eabb4757. [PMID: 32718991 DOI: 10.1126/scitranslmed.abb4757] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022]
Abstract
Vaccine development has the potential to be accelerated by coupling tools such as systems immunology analyses and controlled human infection models to define the protective efficacy of prospective immunogens without expensive and slow phase 2b/3 vaccine studies. Among human challenge models, controlled human malaria infection trials have long been used to evaluate candidate vaccines, and RTS,S/AS01 is the most advanced malaria vaccine candidate, reproducibly demonstrating 40 to 80% protection in human challenge studies in malaria-naïve individuals. Although antibodies are critical for protection after RTS,S/AS01 vaccination, antibody concentrations are inconsistently associated with protection across studies, and the precise mechanism(s) by which vaccine-induced antibodies provide protection remains enigmatic. Using a comprehensive systems serological profiling platform, the humoral correlates of protection against malaria were identified and validated across multiple challenge studies. Rather than antibody concentration, qualitative functional humoral features robustly predicted protection from infection across vaccine regimens. Despite the functional diversity of vaccine-induced immune responses across additional RTS,S/AS01 vaccine studies, the same antibody features, antibody-mediated phagocytosis and engagement of Fc gamma receptor 3A (FCGR3A), were able to predict protection across two additional human challenge studies. Functional validation using monoclonal antibodies confirmed the protective role of Fc-mediated antibody functions in restricting parasite infection both in vitro and in vivo, suggesting that these correlates may mechanistically contribute to parasite restriction and can be used to guide the rational design of an improved vaccine against malaria.
Collapse
Affiliation(s)
- Todd J Suscovich
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | | | - Jishnu Das
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Allison R Demas
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jonathan Crain
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Caitlyn H Linde
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Ashlin Michell
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Harini Natarajan
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Claudia Arevalo
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Thomas Broge
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Thomas Linnekin
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Viraj Kulkarni
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Richard Lu
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Matthew D Slein
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | | | - Meghan Marquette
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Scott Gregory
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | | | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Elke Bergmann-Leitner
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jenny Hendriks
- Janssen Vaccines & Prevention B.V., 2333CN Leiden, Netherlands
| | - Jerald Sadoff
- Janssen Vaccines & Prevention B.V., 2333CN Leiden, Netherlands
| | - Sheetij Dutta
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA.,Broad Institute, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Douglas A Lauffenburger
- Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | - Galit Alter
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
5
|
Roman FP, Coccia M, Schuerman L. Commentary in reply to a publication on Plasmodium falciparum pre-erythrocytic stage vaccine development. Malar J 2020; 19:261. [PMID: 32690097 PMCID: PMC7370254 DOI: 10.1186/s12936-020-03334-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/13/2020] [Indexed: 11/10/2022] Open
Abstract
We have read the publication of Molina-Franky and colleagues on Plasmodium falciparum pre-erythrocytic stage vaccine development (Malaria Journal, 2020;19:56). The commentary revises some of their statements on the RTS,S/AS01 vaccine that are considered either imprecise or incorrect.
Collapse
|
6
|
Dawson L, Earl J, Livezey J. Severe Acute Respiratory Syndrome Coronavirus 2 Human Challenge Trials: Too Risky, Too Soon. J Infect Dis 2020; 222:514-516. [PMID: 32496536 PMCID: PMC7313940 DOI: 10.1093/infdis/jiaa314] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/03/2020] [Indexed: 01/23/2023] Open
Affiliation(s)
- Liza Dawson
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Jake Earl
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Jeffrey Livezey
- Department of Pediatrics, Uniformed Services University of the Health Sciences
| |
Collapse
|
7
|
Pallikkuth S, Chaudhury S, Lu P, Pan L, Jongert E, Wille-Reece U, Pahwa S. A delayed fractionated dose RTS,S AS01 vaccine regimen mediates protection via improved T follicular helper and B cell responses. eLife 2020; 9:51889. [PMID: 32342859 PMCID: PMC7213985 DOI: 10.7554/elife.51889] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 04/14/2020] [Indexed: 12/15/2022] Open
Abstract
Malaria-071, a controlled human malaria infection trial, demonstrated that administration of three doses of RTS,S/AS01 malaria vaccine given at one-month intervals was inferior to a delayed fractional dose (DFD) schedule (62.5% vs 86.7% protection, respectively). To investigate the underlying immunologic mechanism, we analyzed the B and T peripheral follicular helper cell (pTfh) responses. Here, we show that protection in both study arms was associated with early induction of functional IL-21-secreting circumsporozoite (CSP)-specific pTfh cells, together with induction of CSP-specific memory B cell responses after the second dose that persisted after the third dose. Data integration of key immunologic measures identified a subset of non-protected individuals in the standard (STD) vaccine arm who lost prior protective B cell responses after receiving the third vaccine dose. We conclude that the DFD regimen favors persistence of functional B cells after the third dose.
Collapse
Affiliation(s)
- Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, United States
| | - Sidhartha Chaudhury
- Biotechnology HPC Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, United States
| | - Pinyi Lu
- Biotechnology HPC Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, United States.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Rockville, United States
| | - Li Pan
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, United States
| | | | | | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, United States
| |
Collapse
|
8
|
Abstract
Following successful phase 1 and 2 trials, RTS,S/AS01 was evaluated in a large phase 3 trial in 7 African countries in which 8922 young children and 6537 infants were enrolled and followed for a median of 48 and 38 months, respectively. The vaccine efficacy against uncomplicated malaria in children was 28% without booster and 36% with booster. The vaccine received the approval of the European Medical Agency in 2015, but two WHO expert committees requested more data before the programmatic use of RTS,S/AS01 in African children can be recommended. To provide such data, a very large, cluster randomized trial is currently in progress in three African countries. The integration of RTS,S/AS01 in national childhood vaccination programs will benefit the individual vaccinated child but is unlikely to make an impact on malaria transmission. Mass vaccination campaigns that include all age groups over short periods are more likely to harness the short but high protection afforded by RTS,S/AS01.
Collapse
Affiliation(s)
- Lorenz von Seidlein
- Mahidol-Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
9
|
McCall MBB, Kremsner PG, Mordmüller B. Correlating efficacy and immunogenicity in malaria vaccine trials. Semin Immunol 2018; 39:52-64. [PMID: 30219621 DOI: 10.1016/j.smim.2018.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 12/19/2022]
Abstract
The availability of an effective and appropriately implemented malaria vaccine would form a crucial cornerstone of public health efforts to fight this disease. Despite many decades of research, however, no malaria vaccine has yet shown satisfactory protective efficacy or been rolled-out. Validated immunological substitute endpoints have the potential to accelerate clinical vaccine development by reducing the required complexity, size, duration and cost of clinical trials. Besides facilitating clinical development of existing vaccine candidates, understanding immunological mechanisms of protection may drive the development of fundamentally new vaccination approaches. In this review we focus on correlates of protection in malaria vaccine development: Does immunogenicity predict malaria vaccine efficacy and why is this question particularly difficult? Have immunological correlates accelerated malaria vaccine development in the past and will they facilitate it in the future? Does Controlled Human Malaria Infection represent a valid model for identifying such immunological correlates, or a correlate of protection against naturally-acquired malaria in itself?
Collapse
Affiliation(s)
- Matthew B B McCall
- Institut für Tropenmedizin, Universität Tübingen and Deutsches Zentrum für Infektionsforschung, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.
| | - Peter G Kremsner
- Institut für Tropenmedizin, Universität Tübingen and Deutsches Zentrum für Infektionsforschung, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Benjamin Mordmüller
- Institut für Tropenmedizin, Universität Tübingen and Deutsches Zentrum für Infektionsforschung, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
10
|
Roestenberg M, Hoogerwerf MA, Ferreira DM, Mordmüller B, Yazdanbakhsh M. Experimental infection of human volunteers. THE LANCET. INFECTIOUS DISEASES 2018; 18:e312-e322. [PMID: 29891332 DOI: 10.1016/s1473-3099(18)30177-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 02/08/2018] [Accepted: 03/01/2018] [Indexed: 12/13/2022]
Abstract
Controlled human infection (CHI) trials, in which healthy volunteers are experimentally infected, can accelerate the development of novel drugs and vaccines for infectious diseases of global importance. The use of CHI models is expanding from around 60 studies in the 1970s to more than 120 publications in this decade, primarily for influenza, rhinovirus, and malaria. CHI trials have provided landmark data for several registered drugs and vaccines, and have generated unprecedented scientific insights. Because of their invasive nature, CHI studies demand critical ethical review according to established frameworks. CHI-associated serious adverse events are rarely reported. Novel CHI models need standardised safety data from comparable CHI models to facilitate evidence-based risk assessments, as well as funds to produce challenge inoculum according to regulatory requirements. Advances such as the principle of controlled colonisation, the expansion of models to endemic areas, and the use of genetically attenuated strains will further broaden the scope of CHI trials.
Collapse
Affiliation(s)
| | | | | | - Benjamin Mordmüller
- Institute of Tropical Medicine and German Center for Infection Research, partner site Tübingen, University of Tübingen, Tübingen, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | |
Collapse
|
11
|
Najer A, Palivan CG, Beck HP, Meier W. Challenges in Malaria Management and a Glimpse at Some Nanotechnological Approaches. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1052:103-112. [PMID: 29785484 DOI: 10.1007/978-981-10-7572-8_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Malaria is a devastating infectious disease transmitted by mosquitoes, affecting millions of people and killing about half a million children each year. Despite tremendous progress in the control and elimination of malaria within the past years, there are still considerable challenges to be solved. To name a few, drug-resistant parasites, insecticide-resistant mosquitoes and the difficulty to formulate a potent malaria vaccine need to be addressed with new strategies to achieve the final goal of malaria eradication. Nanotechnology-researching and designing innovative structures at the nanoscale-is a promising contemporary technology that is being applied to a vast number of biomedical problems. In the case of malaria, nanotechnology provides tools to design strategies to target drug molecules to specific stages of the parasite, treat drug-resistant parasites, resolve severe malaria, increase vaccine efficacies and combinations thereof. This chapter introduces malaria, discusses current challenges of malaria control and relates these challenges to some potential solutions provided by the nanotechnology field.
Collapse
Affiliation(s)
- Adrian Najer
- Department of Chemistry, University of Basel, 4056, Basel, Switzerland.,Swiss Tropical and Public Health Institute, University of Basel, 4002, Basel, Switzerland
| | | | - Hans-Peter Beck
- Swiss Tropical and Public Health Institute, University of Basel, 4002, Basel, Switzerland
| | - Wolfgang Meier
- Department of Chemistry, University of Basel, 4056, Basel, Switzerland.
| |
Collapse
|
12
|
Mbengue B, Kpodji P, Sylla Niang M, Varela ML, Thiam A, Sow A, Ndiaye K, Aidara M, Thiam F, Ndiaye R, Diop G, Nguer CM, Perraut R, Dièye A. [Profiles of IgG responses against CSP, GLURP and LSA-3NR2 in urban malaria (Dakar): relations with haemoglobin levels and parasite densities]. BULLETIN DE LA SOCIETE DE PATHOLOGIE EXOTIQUE (1990) 2016; 109:91-98. [PMID: 27100862 DOI: 10.1007/s13149-016-0485-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/23/2016] [Indexed: 06/05/2023]
Abstract
Malaria remains a major health problem in sub- Saharan African countries despite substantial decreases in morbidity and mortality due to sustained control programs. Vaccines candidates were mainly tested in rural endemic setting; however increasing proportion of the population is living in urban area. Evaluation of the qualitative or quantitative immune responses to key targets of anti-Plasmodium immunity requires further investigation in urban area. In a cohort of 144 patients with mild malaria living in Dakar, we analyzed IgG responses against target antigens of P. falciparum: CSP, LSA-3NR2 and GLURP by ELISA. A mean age of 15 yrs (4-65 yrs) was found and patients were separated in 59 adults (<15yrs) and 85 children (≤15 yrs). Parasites densities (0,01-15%) did not differ between the two age groups. In contrast, haemoglobin levels appeared lower in children (4.5-16.6 g/dl) (p<0.01). For the immune results, the most recognized antigens were GLURP and CSP compared to LSA-3NR2. Levels of IgG against these antigens were significantly different between the two age groups and they were positively correlated (rho = 0.32; p<0.001). In addition, levels of IgG anti-GLURP were associated with low parasitemia (≤1%) and absence of anemia (≥11g/dl), particularly in adults (p<0.001). In a multiple regression analysis, no significant relationship was found between parasite densities and IgG responses against all the tested antigens. Our study shows the implication of IgG anti-GLURP in humoral immune response against the parasite. The present work contributes to determine IgG levels that can be used as relevant immunologic biomarkers in urban clinical malaria.
Collapse
Affiliation(s)
- B Mbengue
- Service d'immunologie FMPO, Université Cheikh Anta Diop, Dakar, Sénégal.
- Unité d'immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal.
| | - P Kpodji
- Unité d'immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal
| | - M Sylla Niang
- Service d'immunologie FMPO, Université Cheikh Anta Diop, Dakar, Sénégal
| | - M L Varela
- Unité d'immunologie, Institut Pasteur de Dakar, Dakar, Sénégal
| | - A Thiam
- Unité d'immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal
| | - A Sow
- Unité d'immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal
| | - K Ndiaye
- Unité d'immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal
| | - M Aidara
- Unité d'immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal
| | - F Thiam
- Unité d'immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal
| | - R Ndiaye
- Unité d'immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal
| | - G Diop
- Unité d'immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal
| | - C M Nguer
- Département génie chimique et biologie appliquée, ESP, Université Cheikh Anta Diop, Dakar, Sénégal
| | - R Perraut
- Unité d'immunologie, Institut Pasteur de Dakar, Dakar, Sénégal
| | - A Dièye
- Unité d'immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal
| |
Collapse
|
13
|
Ladd Effio C, Oelmeier SA, Hubbuch J. High-throughput characterization of virus-like particles by interlaced size-exclusion chromatography. Vaccine 2016; 34:1259-67. [PMID: 26845741 DOI: 10.1016/j.vaccine.2016.01.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 01/12/2016] [Accepted: 01/17/2016] [Indexed: 11/26/2022]
Abstract
The development and manufacturing of safe and effective vaccines relies essentially on the availability of robust and precise analytical techniques. Virus-like particles (VLPs) have emerged as an important and valuable class of vaccines for the containment of infectious diseases. VLPs are produced by recombinant protein expression followed by purification procedures to minimize the levels of process- and product-related impurities. The control of these impurities is necessary during process development and manufacturing. Especially monitoring of the VLP size distribution is important for the characterization of the final vaccine product. Currently used methods require long analysis times and tailor-made assays. In this work, we present a size-exclusion ultra-high performance liquid chromatography (SE-UHPLC) method to characterize VLPs and quantify aggregates within 3.1min per sample applying interlaced injections. Four analytical SEC columns were evaluated for the analysis of human B19 parvo-VLPs and murine polyoma-VLPs. The optimized method was successfully used for the characterization of five recombinant protein-based VLPs including human papillomavirus (HPV) VLPs, human enterovirus 71 (EV71) VLPs, and chimeric hepatitis B core antigen (HBcAg) VLPs pointing out the generic applicability of the assay. Measurements were supported by transmission electron microscopy and dynamic light scattering. It was demonstrated that the iSE-UHPLC method provides a rapid, precise and robust tool for the characterization of VLPs. Two case studies on purification tools for VLP aggregates and storage conditions of HPV VLPs highlight the relevance of the analytical method for high-throughput process development and process monitoring of virus-like particles.
Collapse
Affiliation(s)
- Christopher Ladd Effio
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany
| | - Stefan A Oelmeier
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany; Boehringer Ingelheim Pharma GmbH & Co. KG, Germany
| | - Jürgen Hubbuch
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany.
| |
Collapse
|
14
|
Ladd Effio C, Baumann P, Weigel C, Vormittag P, Middelberg A, Hubbuch J. High-throughput process development of an alternative platform for the production of virus-like particles in Escherichia coli. J Biotechnol 2015; 219:7-19. [PMID: 26707548 DOI: 10.1016/j.jbiotec.2015.12.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/17/2015] [Accepted: 12/14/2015] [Indexed: 11/26/2022]
Abstract
The production of safe vaccines against untreatable or new diseases has pushed the research in the field of virus-like particles (VLPs). Currently, a large number of commercial VLP-based human vaccines and vaccine candidates are available or under development. A promising VLP production route is the controlled in vitro assembly of virus proteins into capsids. In the study reported here, a high-throughput screening (HTS) procedure was implemented for the upstream process development of a VLP platform in bacterial cell systems. Miniaturized cultivations were carried out in 48-well format in the BioLector system (m2p-Labs, Germany) using an Escherichia coli strain with a tac promoter producing the murine polyomavirus capsid protein (VP1). The screening procedure incorporated micro-scale cultivations, HTS cell disruption by sonication and HTS-compatible analytics by capillary gel electrophoresis. Cultivation temperatures, shaking speeds, induction and medium conditions were varied to optimize the product expression in E. coli. The most efficient system was selected based on an evaluation of soluble and insoluble product concentrations as well as on the percentage of product in the total soluble protein fraction. The optimized system was scaled up to cultivation 2.5L shaker flask scale and purified using an anion exchange chromatography membrane adsorber, followed by a size exclusion chromatography polishing procedure. For proof of concept, purified VP1 capsomeres were assembled under defined buffer conditions into empty capsids and characterized using transmission electron microscopy (TEM). The presented HTS procedure allowed for a fast development of an efficient production process of VLPs in E. coli. Under optimized cultivation conditions, the VP1 product totalled up to 43% of the total soluble protein fraction, yielding 1.63 mg VP1 per mL of applied cultivation medium. The developed production process strongly promotes the murine polyoma-VLP platform, moving towards an industrially feasible technology for new chimeric vaccines.
Collapse
Affiliation(s)
- Christopher Ladd Effio
- Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| | - Pascal Baumann
- Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| | - Claudia Weigel
- Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Philipp Vormittag
- Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Anton Middelberg
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Australia
| | - Jürgen Hubbuch
- Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| |
Collapse
|
15
|
Khan F, Porter M, Schwenk R, DeBot M, Saudan P, Dutta S. Head-to-Head Comparison of Soluble vs. Qβ VLP Circumsporozoite Protein Vaccines Reveals Selective Enhancement of NANP Repeat Responses. PLoS One 2015; 10:e0142035. [PMID: 26571021 PMCID: PMC4646581 DOI: 10.1371/journal.pone.0142035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/17/2015] [Indexed: 11/23/2022] Open
Abstract
Circumsporozoite protein (CSP) of Plasmodium falciparum is a promising malaria vaccine target. RTS,S, the most advanced malaria vaccine candidate consists of the central NANP repeat and carboxy-terminal region of CSP displayed on a hepatitis B virus-like particle (VLP). To build upon the success of RTS,S, we produced a near full-length Plasmodium falciparum CSP that also includes the conserved amino-terminal region of CSP. We recently showed that this soluble CSP, combined with a synthetic Toll-like-receptor-4 (TLR4) agonist in stable oil-in-water emulsion (GLA/SE), induces a potent and protective immune response in mice against transgenic parasite challenge. Here we have investigated whether the immunogenicity of soluble CSP could be further augmented by presentation on a VLP. Bacteriophage Qβ VLPs can be readily produced in E.coli, they have a diameter of 25 nm and contain packaged E. coli RNA which serves as a built in adjuvant through the activation of TLR7/8. CSP was chemically conjugated to Qβ and the CSP-Qβ vaccine immunogenicity and efficacy were compared to adjuvanted soluble CSP in the C57Bl/6 mouse model. When formulated with adjuvants lacking a TLR4 agonist (Alum, SE and Montanide) the Qβ-CSP induced higher anti-NANP repeat titers, higher levels of cytophilic IgG2b/c antibodies and a trend towards higher protection against transgenic parasite challenge as compared to soluble CSP formulated in the same adjuvant. The VLP and soluble CSP immunogenicity difference was most pronounced at low antigen dose, and within the CSP molecule, the titers against the NANP repeats were preferentially enhanced by Qβ presentation. While a TLR4 agonist enhanced the immunogenicity of soluble CSP to levels comparable to the VLP vaccine, the TLR4 agonist did not further improve the immunogenicity of the Qβ-CSP vaccine. The data presented here pave the way for further improvement in the Qβ conjugation chemistry and evaluation of both the Qβ-CSP and soluble CSP vaccines in the non-human primate model.
Collapse
Affiliation(s)
- Farhat Khan
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Mike Porter
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Robert Schwenk
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Margot DeBot
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Philippe Saudan
- Cytos Biotechnology, Wagistrasse 25, 8952 Schlieren, Switzerland
| | - Sheetij Dutta
- Structural Vaccinology Laboratory, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
- * E-mail:
| |
Collapse
|
16
|
Effio CL, Hubbuch J. Next generation vaccines and vectors: Designing downstream processes for recombinant protein-based virus-like particles. Biotechnol J 2015; 10:715-27. [PMID: 25880158 DOI: 10.1002/biot.201400392] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/11/2015] [Accepted: 03/19/2015] [Indexed: 12/28/2022]
Abstract
In recent years, the development of novel recombinant virus-like particles (VLPs) has been generating new perspectives for the prevention of untreated and arising infectious diseases. However, cost-reduction and acceleration of manufacturing processes for VLP-based vaccines or vectors are key challenges for the global health system. In particular, the design of rapid and cost-efficient purification processes is a critical bottleneck. In this review, we describe and evaluate new concepts, development strategies and unit operations for the downstream processing of VLPs. A special focus is placed on purity requirements and current trends, as well as chances and limitations of novel technologies. The discussed methods and case studies demonstrate the advances and remaining challenges in both rational process development and purification tools for large biomolecules. The potential of a new era of VLP-based products is highlighted by the progress of various VLPs in clinical phases.
Collapse
Affiliation(s)
- Christopher Ladd Effio
- Karlsruhe Institute of Technology, Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe, Germany
| | | |
Collapse
|
17
|
da Costa M, Pinheiro-Silva R, Antunes S, Moreno-Cid JA, Custódio A, Villar M, Silveira H, de la Fuente J, Domingos A. Mosquito Akirin as a potential antigen for malaria control. Malar J 2014; 13:470. [PMID: 25472895 PMCID: PMC4265507 DOI: 10.1186/1475-2875-13-470] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/27/2014] [Indexed: 11/10/2022] Open
Abstract
Background The control of vector-borne diseases is important to improve human and animal health worldwide. Malaria is one of the world’s deadliest diseases and is caused by protozoan parasites of the genus Plasmodium, which are transmitted by Anopheles spp. mosquitoes. Recent evidences using Subolesin (SUB) and Akirin (AKR) vaccines showed a reduction in the survival and/or fertility of blood-sucking ectoparasite vectors and the infection with vector-borne pathogens. These experiments suggested the possibility of using AKR for malaria control. Methods The role of AKR on Plasmodium berghei infection and on the fitness and reproduction of the main malaria vector, Anopheles gambiae was characterized by evaluating the effect of akr gene knockdown or vaccination with recombinant mosquito AKR on parasite infection levels, fertility and mortality of female mosquitoes. Results Gene knockdown by RNA interference in mosquitoes suggested a role for akr in mosquito survival and fertility. Vaccination with recombinant Aedes albopictus AKR reduced parasite infection in mosquitoes fed on immunized mice when compared to controls. Conclusions These results showed that recombinant AKR could be used to develop vaccines for malaria control. If effective, AKR-based vaccines could be used to immunize wildlife reservoir hosts and/or humans to reduce the risk of pathogen transmission. However, these vaccines need to be evaluated under field conditions to characterize their effect on vector populations and pathogen infection and transmission.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ana Domingos
- Instituto de Higiene e Medicina Tropical, Rua da Junqueira 100, 1349-008 Lisbon, Portugal.
| |
Collapse
|
18
|
Birkett AJ, Moorthy VS, Loucq C, Chitnis CE, Kaslow DC. Malaria vaccine R&D in the Decade of Vaccines: breakthroughs, challenges and opportunities. Vaccine 2014; 31 Suppl 2:B233-43. [PMID: 23598488 DOI: 10.1016/j.vaccine.2013.02.040] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 02/06/2013] [Accepted: 02/25/2013] [Indexed: 01/23/2023]
Abstract
While recent progress has been made in reducing malaria mortality with other interventions, vaccines are still urgently needed to further reduce the incidence of clinical disease, including during pregnancy, and to provide "herd protection" by blocking parasite transmission. The most clinically advanced candidate, RTS,S, is presently undergoing Phase 3 evaluation in young African children across 13 clinical sites in eight African countries. In the 12-month period following vaccination, RTS,S conferred approximately 50% protection from clinical Plasmodium falciparum disease in children aged 5-17 months, and approximately 30% protection in children aged 6-12 weeks when administered in conjunction with Expanded Program for Immunization (EPI) vaccines. The development of more highly efficacious vaccines to prevent clinical disease caused by both P. falciparum and Plasmodium vivax, as well as vaccines to support elimination efforts by inducing immunity that blocks malaria parasite transmission, are priorities. Some key barriers to malaria vaccine development include: a paucity of well-characterized target immunogens and an absence of clear correlates of protection to enable vaccine development targeting all stages of the P. falciparum and P. vivax lifecycles; a limited number of safe and effective delivery systems, including adjuvants, that induce potent, long-lived protective immunity, be it by antibody, CD4+, and/or CD8+ T cell responses; and, for vaccines designed to provide "herd protection" by targeting sexual stage and/or mosquito antigens, the lack of a clear clinical and regulatory pathway to licensure using non-traditional endpoints. Recommendations to overcome these, and other key challenges, are suggested in this document.
Collapse
Affiliation(s)
- Ashley J Birkett
- PATH Malaria Vaccine Initiative, Washington, DC 20001-2621, USA.
| | | | | | | | | |
Collapse
|
19
|
Inducing humoral and cellular responses to multiple sporozoite and liver-stage malaria antigens using exogenous plasmid DNA. Infect Immun 2013; 81:3709-20. [PMID: 23897618 DOI: 10.1128/iai.00180-13] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A vaccine candidate that elicits humoral and cellular responses to multiple sporozoite and liver-stage antigens may be able to confer protection against Plasmodium falciparum malaria; however, a technology for formulating and delivering such a vaccine has remained elusive. Here, we report the preclinical assessment of an optimized DNA vaccine approach that targets four P. falciparum antigens: circumsporozoite protein (CSP), liver stage antigen 1 (LSA1), thrombospondin-related anonymous protein (TRAP), and cell-traversal protein for ookinetes and sporozoites (CelTOS). Synthetic DNA sequences were designed for each antigen with modifications to improve expression and were delivered using in vivo electroporation (EP). Immunogenicity was evaluated in mice and nonhuman primates (NHPs) and assessed by enzyme-linked immunosorbent assay (ELISA), gamma interferon (IFN-γ) enzyme-linked immunosorbent spot (ELISpot) assay, and flow cytometry. In mice, DNA with EP delivery induced antigen-specific IFN-γ production, as measured by ELISpot assay and IgG seroconversion against all antigens. Sustained production of IFN-γ, interleukin-2, and tumor necrosis factor alpha was elicited in both the CD4(+) and CD8(+) T cell compartments. Furthermore, hepatic CD8(+) lymphocytes produced LSA1-specific IFN-γ. The immune responses conferred to mice by this approach translated to the NHP model, which showed cellular responses by ELISpot assay and intracellular cytokine staining. Notably, antigen-specific CD8(+) granzyme B(+) T cells were observed in NHPs. Collectively, the data demonstrate that delivery of gene sequences by DNA/EP encoding malaria parasite antigens is immunogenic in animal models and can harness both the humoral and cellular arms of the immune system.
Collapse
|
20
|
Transgenic parasites stably expressing full-length Plasmodium falciparum circumsporozoite protein as a model for vaccine down-selection in mice using sterile protection as an endpoint. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:803-10. [PMID: 23536694 DOI: 10.1128/cvi.00066-13] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Circumsporozoite protein (CSP) of Plasmodium falciparum is a protective human malaria vaccine candidate. There is an urgent need for models that can rapidly down-select novel CSP-based vaccine candidates. In the present study, the mouse-mosquito transmission cycle of a transgenic Plasmodium berghei malaria parasite stably expressing a functional full-length P. falciparum CSP was optimized to consistently produce infective sporozoites for protection studies. A minimal sporozoite challenge dose was established, and protection was defined as the absence of blood-stage parasites 14 days after intravenous challenge. The specificity of protection was confirmed by vaccinating mice with multiple CSP constructs of differing lengths and compositions. Constructs that induced high NANP repeat-specific antibody titers in enzyme-linked immunosorbent assays were protective, and the degree of protection was dependent on the antigen dose. There was a positive correlation between antibody avidity and protection. The antibodies in the protected mice recognized the native CSP on the parasites and showed sporozoite invasion inhibitory activity. Passive transfer of anti-CSP antibodies into naive mice also induced protection. Thus, we have demonstrated the utility of a mouse efficacy model to down-select human CSP-based vaccine formulations.
Collapse
|
21
|
Brooks A, Ba-Nguz A. Country planning for health interventions under development: lessons from the malaria vaccine decision-making framework and implications for other new interventions. Health Policy Plan 2012; 27 Suppl 2:ii50-61. [PMID: 22513733 PMCID: PMC3328760 DOI: 10.1093/heapol/czs039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Traditionally it has taken years or decades for new public health interventions targeting diseases found in developing countries to be accessible to those most in need. One reason for the delay has been insufficient anticipation of the eventual processes and evidence required for decision making by countries. This paper describes research into the anticipated processes and data needed to inform decision making on malaria vaccines, the most advanced of which is still in phase 3 trials. From 2006 to 2008, a series of country consultations in Africa led to the development of a guide to assist countries in preparing their malaria vaccine decision-making frameworks. The guide builds upon the World Health Organization's Vaccine Introduction Guidelines. It identifies the processes and data for decisions, when they would be needed relative to the development timelines of the intervention, and where they will come from. Policy development will be supported by data (e.g. malaria disease burden; roles of other malaria interventions; malaria vaccine impact; economic and financial issues; malaria vaccine efficacy, quality and safety) as will implementation decisions (e.g. programmatic issues and socio-cultural environment). This generic guide can now be applied to any future malaria vaccine. The paper discusses the opportunities and challenges to early planning for country decision-making-from the potential for timely, evidence-informed decisions to the risks of over-promising around an intervention still under development. Careful and well-structured planning by countries is an important way to ensure that new interventions do not remain unused for years or decades after they become available.
Collapse
Affiliation(s)
- Alan Brooks
- PATH Malaria Vaccine Initiative, Ferney-Voltaire, France
| | | |
Collapse
|
22
|
Brooks A, Nunes JK, Garnett A, Biellik R, Leboulleux D, Birkett AJ, Loucq C. Aligning new interventions with developing country health systems: target product profiles, presentation, and clinical trial design. Glob Public Health 2012; 7:931-45. [PMID: 22783872 PMCID: PMC3479626 DOI: 10.1080/17441692.2012.699088] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 04/20/2012] [Indexed: 11/22/2022]
Abstract
Many new interventions are being created to address health problems of the developing world. However, many developing countries have fragile health systems and find it difficult to accommodate change. Consequently, it is essential that new interventions are well aligned with health systems and their users. Establishing target product profiles (TPPs) is a critical, early step towards tailoring interventions to suit both of these constituencies. Specific analyses can help identify and establish relevant TPP criteria such as optimal formulation, presentation and packaging. Clinical trials for a new intervention should be designed to address both TPP-specific questions and anticipated use of the intervention in target countries. Examples are provided from research on malaria vaccines that are also applicable to other new public health interventions.
Collapse
Affiliation(s)
- Alan Brooks
- PATH Malaria Vaccine Initiative, Ferney- Voltaire, France
- Epidemiology and Public Health Department, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
23
|
Horowitz A, Hafalla JCR, King E, Lusingu J, Dekker D, Leach A, Moris P, Cohen J, Vekemans J, Villafana T, Corran PH, Bejon P, Drakeley CJ, von Seidlein L, Riley EM. Antigen-specific IL-2 secretion correlates with NK cell responses after immunization of Tanzanian children with the RTS,S/AS01 malaria vaccine. THE JOURNAL OF IMMUNOLOGY 2012; 188:5054-62. [PMID: 22504653 DOI: 10.4049/jimmunol.1102710] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RTS,S/AS01, a vaccine targeting pre-erythrocytic stages of Plasmodium falciparum, is undergoing clinical trials. We report an analysis of cellular immune response to component Ags of RTS,S-hepatitis B surface Ag (HBs) and P. falciparum circumsporozoite (CS) protein-among Tanzanian children in a phase IIb RTS,S/AS01(E) trial. RTS,S/AS01 (E) vaccinees make stronger T cell IFN-γ, CD69, and CD25 responses to HBs peptides than do controls, indicating that RTS,S boosts pre-existing HBs responses. T cell CD69 and CD25 responses to CS and CS-specific secreted IL-2 were augmented by RTS,S vaccination. Importantly, more than 50% of peptide-induced IFN-γ(+) lymphocytes were NK cells, and the magnitude of the NK cell CD69 response to HBs peptides correlated with secreted IL-2 concentration. CD69 and CD25 expression and IL-2 secretion may represent sensitive markers of RTS,S-induced, CS-specific T cells. The potential for T cell-derived IL-2 to augment NK cell activation in RTS,S-vaccinated individuals, and the relevance of this for protection, needs to be explored further.
Collapse
Affiliation(s)
- Amir Horowitz
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Brooks A, Briët OJT, Hardy D, Steketee R, Smith TA. Simulated impact of RTS,S/AS01 vaccination programs in the context of changing malaria transmission. PLoS One 2012; 7:e32587. [PMID: 22412892 PMCID: PMC3295753 DOI: 10.1371/journal.pone.0032587] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 02/01/2012] [Indexed: 12/29/2022] Open
Abstract
Introduction The RTS,S/AS01 pre-erythrocytic malaria vaccine is in phase III clinical trials. It is critical to anticipate where and how it should be implemented if trials are successful. Such planning may be complicated by changing levels of malaria transmission. Methods/results Computer simulations were used to examine RTS,S/AS01 impact, using a vaccine profile based on phase II trial results, and assuming that protection decays only slowly. Settings were simulated in which baseline transmission (in the absence of vaccine) was fixed or varied between 2 and 20 infectious mosquito bites per person per annum (ibpa) over ten years. Four delivery strategies were studied: routine infant immunization (EPI), EPI plus infant catch-up, EPI plus school-based campaigns, and EPI plus mass campaigns. Impacts in changing transmission settings were similar to those in fixed settings. Assuming a persistent effect of vaccination, at 2 ibpa, the vaccine averted approximately 5–7 deaths per 1000 doses of vaccine when delivered via mass campaigns, but the benefit was less at higher transmission levels. EPI, catch-up and school-based strategies averted 2–3 deaths per 1000 doses in settings with 2 ibpa. In settings where transmission was decreasing or increasing, EPI, catch-up and school-based strategies averted approximately 3–4 deaths per 1000 doses. Discussion Where transmission is changing, it appears to be sufficient to consider simulations of pre-erythrocytic vaccine impact at a range of initial transmission levels. At 2 ibpa, mass campaigns averted the most deaths and reduced transmission, but this requires further study. If delivered via EPI, RTS,S/AS01 could avert approximately 6–11 deaths per 1000 vaccinees in all examined settings, similar to estimates for pneumococcal conjugate vaccine in African infants. These results support RTS,S/AS01 implementation via EPI, for example alongside vector control interventions, providing that the phase III trials provide support for our assumptions about efficacy.
Collapse
Affiliation(s)
- Alan Brooks
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland.
| | | | | | | | | |
Collapse
|
25
|
Expression, immunogenicity, histopathology, and potency of a mosquito-based malaria transmission-blocking recombinant vaccine. Infect Immun 2012; 80:1606-14. [PMID: 22311924 DOI: 10.1128/iai.06212-11] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Vaccines have been at the forefront of global research efforts to combat malaria, yet despite several vaccine candidates, this goal has yet to be realized. A potentially effective approach to disrupting the spread of malaria is the use of transmission-blocking vaccines (TBV), which prevent the development of malarial parasites within their mosquito vector, thereby abrogating the cascade of secondary infections in humans. Since malaria is transmitted to human hosts by the bite of an obligate insect vector, mosquito species in the genus Anopheles, targeting mosquito midgut antigens that serve as ligands for Plasmodium parasites represents a promising approach to breaking the transmission cycle. The midgut-specific anopheline alanyl aminopeptidase N (AnAPN1) is highly conserved across Anopheles vectors and is a putative ligand for Plasmodium ookinete invasion. We have developed a scalable, high-yield Escherichia coli expression and purification platform for the recombinant AnAPN1 TBV antigen and report on its marked vaccine potency and immunogenicity, its capacity for eliciting transmission-blocking antibodies, and its apparent lack of immunization-associated histopathologies in a small-animal model.
Collapse
|
26
|
Superinfection in malaria: Plasmodium shows its iron will. EMBO Rep 2011; 12:1233-42. [PMID: 22081142 DOI: 10.1038/embor.2011.213] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 09/30/2011] [Indexed: 12/21/2022] Open
Abstract
After the bite of a malaria-infected mosquito, the Plasmodium sporozoite infects liver cells and produces thousands of merozoites, which then infect red blood cells, causing malaria. In malaria-endemic areas, several hundred infected mosquitoes can bite an individual each year, increasing the risk of superinfection. However, in infants that are yet to acquire immunity, superinfections are infrequent. We have recently shown that blood-stage parasitaemia, above a minimum threshold, impairs the growth of a subsequent sporozoite infection of liver cells. Blood-stage parasites stimulate the production of the host iron-regulatory factor hepcidin, which redistributes iron away from hepatocytes, reducing the development of the iron-dependent liver stage. This could explain why Plasmodium superinfection is not often found in young nonimmune children. Here, we discuss the impact that such protection from superinfection might have in epidemiological settings or in programmes for controlling malaria, as well as how the induction of hepcidin and redistribution of iron might influence anaemia and the outcome of non-Plasmodium co-infections.
Collapse
|
27
|
Agnandji ST, Lell B, Soulanoudjingar SS, Fernandes JF, Abossolo BP, Conzelmann C, Methogo BGNO, Doucka Y, Flamen A, Mordmüller B, Issifou S, Kremsner PG, Sacarlal J, Aide P, Lanaspa M, Aponte JJ, Nhamuave A, Quelhas D, Bassat Q, Mandjate S, Macete E, Alonso P, Abdulla S, Salim N, Juma O, Shomari M, Shubis K, Machera F, Hamad AS, Minja R, Mtoro A, Sykes A, Ahmed S, Urassa AM, Ali AM, Mwangoka G, Tanner M, Tinto H, D'Alessandro U, Sorgho H, Valea I, Tahita MC, Kaboré W, Ouédraogo S, Sandrine Y, Guiguemdé RT, Ouédraogo JB, Hamel MJ, Kariuki S, Odero C, Oneko M, Otieno K, Awino N, Omoto J, Williamson J, Muturi-Kioi V, Laserson KF, Slutsker L, Otieno W, Otieno L, Nekoye O, Gondi S, Otieno A, Ogutu B, Wasuna R, Owira V, Jones D, Onyango AA, Njuguna P, Chilengi R, Akoo P, Kerubo C, Gitaka J, Maingi C, Lang T, Olotu A, Tsofa B, Bejon P, Peshu N, Marsh K, Owusu-Agyei S, Asante KP, Osei-Kwakye K, Boahen O, Ayamba S, Kayan K, Owusu-Ofori R, Dosoo D, Asante I, Adjei G, Adjei G, Chandramohan D, Greenwood B, Lusingu J, Gesase S, Malabeja A, Abdul O, Kilavo H, Mahende C, Liheluka E, Lemnge M, Theander T, Drakeley C, Ansong D, Agbenyega T, Adjei S, Boateng HO, Rettig T, Bawa J, Sylverken J, Sambian D, Agyekum A, Owusu L, Martinson F, Hoffman I, Mvalo T, Kamthunzi P, Nkomo R, Msika A, Jumbe A, Chome N, Nyakuipa D, Chintedza J, Ballou WR, Bruls M, Cohen J, Guerra Y, Jongert E, Lapierre D, Leach A, Lievens M, Ofori-Anyinam O, Vekemans J, Carter T, Leboulleux D, Loucq C, Radford A, Savarese B, Schellenberg D, Sillman M, Vansadia P. First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. N Engl J Med 2011; 365:1863-75. [PMID: 22007715 DOI: 10.1056/nejmoa1102287] [Citation(s) in RCA: 625] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND An ongoing phase 3 study of the efficacy, safety, and immunogenicity of candidate malaria vaccine RTS,S/AS01 is being conducted in seven African countries. METHODS From March 2009 through January 2011, we enrolled 15,460 children in two age categories--6 to 12 weeks of age and 5 to 17 months of age--for vaccination with either RTS,S/AS01 or a non-malaria comparator vaccine. The primary end point of the analysis was vaccine efficacy against clinical malaria during the 12 months after vaccination in the first 6000 children 5 to 17 months of age at enrollment who received all three doses of vaccine according to protocol. After 250 children had an episode of severe malaria, we evaluated vaccine efficacy against severe malaria in both age categories. RESULTS In the 14 months after the first dose of vaccine, the incidence of first episodes of clinical malaria in the first 6000 children in the older age category was 0.32 episodes per person-year in the RTS,S/AS01 group and 0.55 episodes per person-year in the control group, for an efficacy of 50.4% (95% confidence interval [CI], 45.8 to 54.6) in the intention-to-treat population and 55.8% (97.5% CI, 50.6 to 60.4) in the per-protocol population. Vaccine efficacy against severe malaria was 45.1% (95% CI, 23.8 to 60.5) in the intention-to-treat population and 47.3% (95% CI, 22.4 to 64.2) in the per-protocol population. Vaccine efficacy against severe malaria in the combined age categories was 34.8% (95% CI, 16.2 to 49.2) in the per-protocol population during an average follow-up of 11 months. Serious adverse events occurred with a similar frequency in the two study groups. Among children in the older age category, the rate of generalized convulsive seizures after RTS,S/AS01 vaccination was 1.04 per 1000 doses (95% CI, 0.62 to 1.64). CONCLUSIONS The RTS,S/AS01 vaccine provided protection against both clinical and severe malaria in African children. (Funded by GlaxoSmithKline Biologicals and the PATH Malaria Vaccine Initiative; RTS,S ClinicalTrials.gov number, NCT00866619 .).
Collapse
|
28
|
Garçon N, Van Mechelen M. Recent clinical experience with vaccines using MPL- and QS-21-containing adjuvant systems. Expert Rev Vaccines 2011; 10:471-86. [PMID: 21506645 DOI: 10.1586/erv.11.29] [Citation(s) in RCA: 240] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The immunostimulants 3-O-desacyl-4'-monophosphoryl lipid A (MPL) and the saponin QS-21 are part of licensed or candidate vaccines. MPL and QS-21 directly affect the innate immune response to orchestrate the quality and intensity of the adaptive immune response to the vaccine antigens. The combination of immunostimulants in different adjuvant formulations forms the basis of Adjuvant Systems (AS) as a way to promote appropriate protective immune responses following vaccination. MPL and aluminum salts are present in AS04, and both MPL and QS-21 are present in AS01 and AS02, which are liposome- and emulsion-based formulations, respectively. The recent clinical performance of AS01-, AS02- and AS04-adjuvanted vaccines will be discussed in the context of the diseases being targeted. The licensing of two AS04-adjuvanted vaccines and the initiation of Phase III trials with an AS01-adjuvanted vaccine demonstrate the potential to develop new or improved human vaccines that contain MPL or MPL and QS-21.
Collapse
|
29
|
Schwenk RJ, Richie TL. Protective immunity to pre-erythrocytic stage malaria. Trends Parasitol 2011; 27:306-14. [PMID: 21435951 DOI: 10.1016/j.pt.2011.02.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 02/14/2011] [Accepted: 02/14/2011] [Indexed: 01/23/2023]
Abstract
The development of a vaccine against malaria is a major research priority given the burden of disease, death and economic loss inflicted upon the tropical world by this parasite. Despite decades of effort, however, a vaccine remains elusive. The best candidate is a subunit vaccine termed RTS,S but this provides only partial protection against clinical disease. This review examines what is known about protective immunity against pre-erythrocytic stage malaria by considering the humoral and T cell-mediated immune responses that are induced by attenuated sporozoites and by the RTS,S vaccine. On the basis of these observations a set of research priorities are defined that are crucial for the development of a vaccine capable of inducing long-lasting and high-grade protection against malaria.
Collapse
Affiliation(s)
- Robert J Schwenk
- US Military Malaria Vaccine Program, Walter Reed Army Institute of Research, Division of Malaria Vaccine Development, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA.
| | | |
Collapse
|
30
|
Arévalo-Herrera M, Vera O, Castellanos A, Céspedes N, Soto L, Corradin G, Herrera S. Preclinical vaccine study of Plasmodium vivax circumsporozoite protein derived-synthetic polypeptides formulated in montanide ISA 720 and montanide ISA 51 adjuvants. Am J Trop Med Hyg 2011; 84:21-7. [PMID: 21292874 DOI: 10.4269/ajtmh.2011.10-0110] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Plasmodium vivax circumsporozoite (CS) protein is a leading malaria vaccine candidate previously assessed in animals and humans. Here, combinations of three synthetic polypeptides corresponding to amino (N), central repeat (R), and carboxyl (C) regions of the CS protein formulated in Montanide ISA 720 or Montanide ISA 51 adjuvants were assessed for immunogenicity in rodents and primates. BALB/c mice and Aotus monkeys were divided into test and control groups and were immunized three times with doses of 50 and 100 μg of vaccine or placebo. Antigen-specific antimalarial antibodies were determined by enzyme-linked immunosorbent assay, immunofluorescent antibody test, and IFN-γ responses by enzyme-linked immunosorbent spot (ELIspot). Both vaccine formulations were highly immunogenic in both species. Mice developed better antibody responses against C and R polypeptides, whereas the N polypeptide was more immunogenic in monkeys. Anti-peptide antibodies remained detectable for several months and recognized native proteins on sporozoites. Differences between Montanide ISA 720 and Montanide ISA 51 formulations were not significant.
Collapse
|
31
|
Lau OS, Ng DWK, Chan WWL, Chang SP, Sun SSM. Production of the 42-kDa fragment of Plasmodium falciparum merozoite surface protein 1, a leading malaria vaccine antigen, in Arabidopsis thaliana seeds. PLANT BIOTECHNOLOGY JOURNAL 2010; 8:994-1004. [PMID: 20444208 DOI: 10.1111/j.1467-7652.2010.00526.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Malaria is widely associated with poverty, and a low-cost vaccine against malaria is highly desirable for implementing comprehensive vaccination programmes in developing countries. Production of malaria antigens in plants is a promising approach, but its development has been hindered by poor expression of the antigens in plant cells. In the present study, we targeted plant seeds as a low-cost vaccine production platform and successfully expressed the Plasmodium falciparum 42-kDa fragment of merozoite surface protein 1 (MSP1₄₂), a leading malaria vaccine candidate, at a high level in transgenic Arabidopsis seeds. We overcame hurdles of transcript and protein instabilities of MSP1₄₂ in plants by synthesizing a plant-optimized MSP1₄₂ cDNA and either targeting the recombinant protein to protein storage vacuoles or fusing it with a stable plant storage protein. An exceptional improvement in MSP1₄₂ expression, from an undetectable level to 5% of total extractable protein, was achieved with these combined strategies. Importantly, the plant-derived MSP1₄₂ maintains its natural antigenicity and can be recognized by immune sera from malaria-infected patients. Our results provide a strong basis for the development of a plant-based, low-cost malaria vaccine.
Collapse
Affiliation(s)
- On Sun Lau
- Department of Biology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | | | | | | | | |
Collapse
|
32
|
Abstract
A comprehensive 5-week vaccinology course was recently held in Paris (1 March-2 April 2010) hosted by the Institut Pasteur, a world-renowned center for science and vaccinology. A total of 25 young scientists from different parts of the world participated and 63 world experts gave lectures in six specific modules that covered all aspects of vaccinology, from basic research to clinical studies. Students also had the opportunity to attend a 2-day "Pandemic Influenza Workshop". This article summarizes the issues discussed during this course and highlights the importance of global access to vaccines.
Collapse
Affiliation(s)
- Emrah Altindiş
- Research Center, Novartis Vaccines and Diagnostics, Via Fiorentina, 1, 53100, Siena, Italy.
| |
Collapse
|
33
|
The Th1 immune response to Plasmodium falciparum circumsporozoite protein is boosted by adenovirus vectors 35 and 26 with a homologous insert. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1687-94. [PMID: 20826614 DOI: 10.1128/cvi.00311-10] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The most advanced malaria vaccine, RTS,S, is comprised of a portion of the Plasmodium falciparum circumsporozoite (CS) protein, fused to and admixed with the hepatitis B virus surface antigen, and an adjuvant [corrected].This vaccine confers short-term protection against malaria infection, with an efficacy of about 50%, and induces particularly B-cell and CD4(+) T-cell responses.In the present study, we tested the hypothesis that the Th1 immune response to CS protein,in particular the CD8(+) T-cell response, which is needed for strong and lasting malaria immunity, is boosted to sustainable levels by adenovirus vectors 35 and 26 with a homologous insert (Ad35.CS/Ad26.CS) [corrected]. In this study, we evaluated immune responses induced with vaccination regimens based on an adjuvant-containing, yeast-produced complete CS protein followed by two recombinant low-seroprevalence adenoviruses expressing P. falciparum CS antigen, Ad35.CS (subgroup B) and Ad26.CS (subgroup D). Our results show that (i) the yeast (Hansenula polymorpha)produced, adjuvanted full-length CS protein is highly potent in inducing high CS-specific humoral responses in mice but produces poor T-cell responses, (ii) the Ad35.CS vector boosts the gamma interferon-positive (IFN-γ(+)) CD8(+) T-cell response induced by the CS protein immunization and shifts the immune response toward the Th1 type, and (iii) a three-component heterologous vaccination comprised of a CS protein prime followed by boosts with Ad35.CS and Ad26.CS elicits an even more robust and sustainable IFN-γ(+) CD8(+) T-cell response than one- or two-component regimens. The Ad35.CS/Ad26.CS combination boosted particularly the IFN-γ(+) and tumor necrosis factor alpha-positive (TNF-α(+)) T cells, confirming the shift of the immune response from the Th2 type to the Th1 type. These results support the notion of first immunizations of infants with an adjuvanted CS protein vaccine, followed by a booster Ad35.CS/Ad26.CS vaccine at a later age, to induce lasting protection against malaria for which the Th1 response and immune memory is required.
Collapse
|
34
|
Sedegah M, Kim Y, Peters B, McGrath S, Ganeshan H, Lejano J, Abot E, Banania G, Belmonte M, Sayo R, Farooq F, Doolan DL, Regis D, Tamminga C, Chuang I, Bruder JT, King CR, Ockenhouse CF, Faber B, Remarque E, Hollingdale MR, Richie TL, Sette A. Identification and localization of minimal MHC-restricted CD8+ T cell epitopes within the Plasmodium falciparum AMA1 protein. Malar J 2010; 9:241. [PMID: 20735847 PMCID: PMC2939619 DOI: 10.1186/1475-2875-9-241] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 08/24/2010] [Indexed: 12/14/2022] Open
Abstract
Background Plasmodium falciparum apical membrane antigen-1 (AMA1) is a leading malaria vaccine candidate antigen that is expressed by sporozoite, liver and blood stage parasites. Since CD8+ T cell responses have been implicated in protection against pre-erythrocytic stage malaria, this study was designed to identify MHC class I-restricted epitopes within AMA1. Methods A recombinant adenovirus serotype 5 vector expressing P. falciparum AMA1 was highly immunogenic when administered to healthy, malaria-naive adult volunteers as determined by IFN-γ ELISpot responses to peptide pools containing overlapping 15-mer peptides spanning full-length AMA1. Computerized algorithms (NetMHC software) were used to predict minimal MHC-restricted 8-10-mer epitope sequences within AMA1 15-mer peptides active in ELISpot. A subset of epitopes was synthesized and tested for induction of CD8+ T cell IFN-γ responses by ELISpot depletion and ICS assays. A 3-dimensional model combining Domains I + II of P. falciparum AMA1 and Domain III of P. vivax AMA1 was used to map these epitopes. Results Fourteen 8-10-mer epitopes were predicted to bind to HLA supertypes A01 (3 epitopes), A02 (4 epitopes), B08 (2 epitopes) and B44 (5 epitopes). Nine of the 14 predicted epitopes were recognized in ELISpot or ELISpot and ICS assays by one or more volunteers. Depletion of T cell subsets confirmed that these epitopes were CD8+ T cell-dependent. A mixture of the 14 minimal epitopes was capable of recalling CD8+ T cell IFN-γ responses from PBMC of immunized volunteers. Thirteen of the 14 predicted epitopes were polymorphic and the majority localized to the more conserved front surface of the AMA1 model structure. Conclusions This study predicted 14 and confirmed nine MHC class I-restricted CD8+ T cell epitopes on AMA1 recognized in the context of seven HLA alleles. These HLA alleles belong to four HLA supertypes that have a phenotypic frequency between 23% - 100% in different human populations.
Collapse
Affiliation(s)
- Martha Sedegah
- USMMVP, Malaria Department, NMRC, Silver Spring, MD 20910, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Pays JF. [A mosquito net for everyone in 2010]. ACTA ACUST UNITED AC 2010; 103:223-9. [PMID: 20652477 DOI: 10.1007/s13149-010-0068-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 05/25/2010] [Indexed: 10/19/2022]
Abstract
At less than two hundred days of the 2010 deadline for the "Roll Back Malaria" initiative which committed itself to reduce by half, before that date, mortality due to malaria in the world and relying on the latest WHO reports pointing out, in Africa, major shortcomings concerning the accessibility to treatment combinations consisting of artemisinin and on the acknowledged fact that an insufficient number of pregnant women receive an intermittent treatment, the author notes that a coverage, so-called universal, with the use of long action insecticide treated mosquito nets has become the Grail of the battle against malaria, with the perverse effects entailed, namely that of blinding realities or throwing discredit on other types of possible interventions that are not consistent with an accounting logic. He also notes that the average figure of estimated deaths due to malaria was at a quasi stagnation in 2008 and that the lives of 34,000 African children of less than 5 years of age saved between 2006 and 2008 was achieved in the context of the reduction in infant mortality resulting from a series of causes among which it is impossible to individualise malaria with certainty. He finally points out that Eritrea, Rwanda, Zambia, São Tome y Principe and the Tanzanian island of Zanzibar which quite regularly serve as showcases to RBM and UNICEF and which report spectacular progress in the field of prevention of malaria accompanied by a parallel reduction in its mortality, are, for different reasons, far from being representative of the totality of African countries and that they should be considered as exceptions rather than examples to be exploited without restraint. On the other hand, the author considers that deluding the grand public into thinking that a few watchwords, slogans and simple, even simplistic, ideas would enable eradicating malaria given that large sums of money are made available, is not quite honest and may finally prove to be dangerous. He warns against the false hopes concerning the impact of coming discoveries on the antimalarial battle often conjured up to take over today's actions, by recalling that a century of scientific, technological and medical progress has not positively translated into any decisive progress in the prevention or treatment of this disease, in spite of the issue of scientific publications on the subject at every 20 minute interval since more than thirty years. Although the author willingly agrees that huge financial means are essential and for a long time to come, he however believes that they would not suffice to enable, in countries with high rates of transmission, the elimination of a disease that is not solely linked to biological, ecological and entomological parameters, but that is also anchored to the economic, societal, social and cultural contexts that are quite often forgotten and on which it is difficult, but essential, to act in order to obtain long lasting results. He recalls, on this occasion, that man, as an individual in relationship with his surroundings, should also be at the heart of the battle on the same footing as the anopheles and the plasmodium, even if this, at times, leads to clashes between medical logic and native social, traditional, popular, scholarly or religious logics. He regrets, similarly and within the spirit of the Abuja declaration, that the educational systems of African countries having a high transmission rate do not play the role they had to assume in the battle against malaria and are not mobilised much better than they are, by including, for example, in the primary and secondary curriculum of public and private schools, on the same footing as the learning of the alphabet or of the multiplication tables, a compulsory adapted instruction in malaria, so that children not only become victims but also "actors in the battle against malaria". The author finally underlines the absolute necessity to rapidly strengthen health care facilities of the most affected countries, particularly in the rural area, and plead that this intensification becomes a true priority financed on its own merits. The Global Malaria Action Plan, that will take over from the RBM and would have the advantage of masking the insufficient results obtained by the latter, would have to face two huge challenges: an ancient but recurring one that concerns, in the context of a world economic crisis, the financing of the battle against malaria and the other one, new and associated with the development of resistance by Plasmodium falciparum to artemisinin, a first- and last-line antimalaria drug.
Collapse
|
36
|
Sullivan D. Uncertainty in mapping malaria epidemiology: implications for control. Epidemiol Rev 2010; 32:175-87. [PMID: 20581219 DOI: 10.1093/epirev/mxq013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Malaria is a location-specific, dynamic infectious disease transmitted by mosquitoes to humans and is influenced by environmental, vector, parasite, and host factors. The principal purposes of malarial epidemiology are 1) to describe the malarial distribution in space and time along with the physical, biologic, and social etiologic factors and 2) to guide control objectives for either modeling impact or measuring progress of control tactics. Mapping malaria and many of its causative factors has been achieved on many different levels from global distribution to biologic quantitative trait localization in humans, parasites, and mosquitoes. Despite these important achievements, a large degree of uncertainty still exists on the annual burden of malarial cases. Accurate, sensitive detection and treatment of asymptomatic reservoirs important to infectious transmission are additional components necessary for future control measures. Presently spurred by the leadership and funding of Bill and Melinda Gates, the malarial community is developing and implementing plans for elimination of malaria. The challenge for malariologists is to digitally integrate and map epidemiologic factors and intervention measures in space and time to target effective, sustainable control alongside research efforts.
Collapse
Affiliation(s)
- David Sullivan
- The Malaria Research Institute, W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|