1
|
Dwivedi A, Scalsky RJ, Harris DG, Stabler TC, Shrestha B, Joshi S, Gandhi C, Munro JB, Ifeonu OO, Ouedraogo A, Tiono AB, Coulibaly D, Ouattara A, Richie TL, Sim BKL, Plowe CV, Lyke KE, Takala-Harrison S, Hoffman SL, Thera MA, Sirima SB, Laurens MB, Silva JC. Protective targets of PfSPZ vaccines identified from whole-genome sieve analysis of isolates from malaria vaccine efficacy trials in West Africa. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.04.25323352. [PMID: 40093207 PMCID: PMC11908318 DOI: 10.1101/2025.03.04.25323352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Identification of antigens targeted by a protective response is a central quest in malaria vaccinology. Whole-genome sieve analysis (SAWG) in samples collected from placebo-controlled field trials of Plasmodium falciparum (Pf) sporozoite (SPZ) vaccines may enable identification of Pf pre-erythrocytic antigens. We applied SAWG to genomic data generated from Pf isolates collected during two field trials measuring the efficacy, in malaria-exposed African adults, of two PfSPZ vaccines. These randomized, double-blind, placebo-controlled trials were conducted in regions of Mali and Burkina Faso characterized by high seasonal transmission, where parasite genetic diversity is high. Genomic sites in which the vaccine allelic state was significantly underrepresented among breakthrough infections in vaccinees relative to placebo recipients were termed "target sites". Protein-coding loci containing target sites that changed amino acids were termed "target loci". The SAWG conducted on clinical trial samples from the Burkina Faso and Mali trials identified 138 and 80 single-copy protein-coding target loci in the Burkinabe and Malian data sets, respectively, with twelve common to both, a number significantly higher than expected (E = 3.9; 99%CI = [0, 9]). Among these was the thrombospondin-related anonymous protein locus, which encodes PfSSP2|TRAP, one of the most abundant and well-characterized pre-erythrocytic stage antigen as well as other genes encoding membrane-associated proteins of unknown function. These results identify SAWG as a potentially powerful tool for identifying protective vaccine antigens in recombining pathogens with large genome size and reveals potential new protective Pf antigens.
Collapse
Affiliation(s)
- Ankit Dwivedi
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Ryan J. Scalsky
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - David G. Harris
- Department of Computer Science, University of Maryland College Park; College Park, MD 20742, USA
| | | | - Biraj Shrestha
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Sudhaunshu Joshi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Chakshu Gandhi
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - James B. Munro
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Olukemi O. Ifeonu
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | | | - Alfred B. Tiono
- Groupe de Recherche Action en Santé; Ouagadougou, Burkina Faso
| | - Drissa Coulibaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako; Bamako, Mali
| | - Amed Ouattara
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | | | | | - Christopher V. Plowe
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Shannon Takala-Harrison
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | | | - Mahamadou A. Thera
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako; Bamako, Mali
| | | | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, MD 21201, USA
| | - Joana C. Silva
- Institute for Genome Sciences, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine; Baltimore, MD 21201, USA
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (NOVA); 1349-008 Lisboa, Portugal
| |
Collapse
|
2
|
Venkatraman N, Silman D, Bellamy D, Stockdale L, Bowyer G, Edwards NJ, Griffiths O, Lopez FR, Powlson J, Mair C, Folegatti PM, Datoo MS, Morter R, Minassian AM, Poulton I, Collins KA, Brod F, Angell-Manning P, Berrie E, Brendish N, Glenn G, Fries L, Baum J, Blagborough AM, Roberts R, Lawrie AM, Angus B, Lewis DJM, Faust SN, Ewer KJ, Hill AVS. R21 in Matrix-M adjuvant in UK malaria-naive adult men and non-pregnant women aged 18-45 years: an open-label, partially blinded, phase 1-2a controlled human malaria infection study. THE LANCET. MICROBE 2025; 6:100867. [PMID: 39805301 DOI: 10.1016/s2666-5247(24)00083-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND R21 is a novel malaria vaccine, composed of a fusion protein of the malaria circumsporozoite protein and hepatitis B surface antigen. Following favourable safety and immunogenicity in a phase 1 study, we aimed to assess the efficacy of R21 administered with Matrix-M (R21/MM) against clinical malaria in adults from the UK who were malaria naive in a controlled human malaria infection study. METHODS In this open-label, partially blinded, phase 1-2A controlled human malaria infection study undertaken in Oxford, Southampton, and London, UK, we tested five novel vaccination regimens of R21/MM. A standard three-dose regimen (groups 1 and 6) was compared with a reduced (fractional) third dose (groups 2 and 5) of R21/MM, concomitant administration with viral vectors ChAd63-MVA expressing ME-TRAP (group 3), and a two-dose R21/MM regimen (group 7). Controlled Human Malaria Infection (CHMI) was delivered by mosquito bite at Imperial College London, London, UK, 3-4 weeks after final vaccination (or 18 months after final vaccination for group 6) alongside unvaccinated controls (groups 4A and 4B). The primary outcome measures were to assess safety of the vaccines in healthy malaria-naive volunteers and the efficacy (occurrence of blood-stage malaria infection) of the different vaccine regimens compared with non-vaccinated controls after CHMI. The trial was registered with ClinicalTrials.gov (NCT02905019). FINDINGS 66 volunteers were enrolled with 59 undergoing subsequent CHMI. All vaccination schedules were well tolerated. The highest level of protection against CHMI was observed in participants receiving the standard three-dose regimen of R21/MM (group 1, nine of 11 volunteers protected) with protection maintained in three of five volunteers re-challenged by CHMI 7·5 months later. Protection against malaria was also seen in group 2, group 3, and group 5 compared with unvaccinated control participants. Total IgG antibody responses to the NANP repeat region of circumsporozoite protein peaked after the third dose of R21/MM in all volunteers and were well maintained to 90 days after challenge. Reducing the third dose did not affect protection or antibody concentrations. INTERPRETATION Our study shows that R21/MM elicits high-level efficacy against clinical malaria in a controlled human infection model of malaria in adults who are malaria naive. These data supported the evaluation of R21/MM in field efficacy trials in the target population of young children in malaria-endemic areas. FUNDING EU Horizon 2020, the UK Medical Research Council, the European Commission, the UK National Institute of Health Research, the Imperial NIHR Clinical Research Facility, the Oxford NIHR Biomedical Research Centre, and the Wellcome Trust.
Collapse
Affiliation(s)
- Navin Venkatraman
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Daniel Silman
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Duncan Bellamy
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Lisa Stockdale
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Georgina Bowyer
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Nick J Edwards
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Oliver Griffiths
- Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Fernando Ramos Lopez
- Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Jonathan Powlson
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Catherine Mair
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Pedro M Folegatti
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Mehreen S Datoo
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Richard Morter
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Angela M Minassian
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK; Department of Biochemistry, University of Oxford, Oxford, UK
| | - Ian Poulton
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Katharine A Collins
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Florian Brod
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | | | - Eleanor Berrie
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, UK
| | - Nathan Brendish
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | | | | | - Jake Baum
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | | | - Rachel Roberts
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Alison M Lawrie
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Brian Angus
- Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - David J M Lewis
- NIHR/Wellcome Trust Imperial Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, London, UK
| | - Saul N Faust
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Katie J Ewer
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Adrian V S Hill
- Jenner Institute, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
3
|
Rappaport AR, Bekerman E, Boucher GR, Sung J, Carr B, Corzo CA, Larson H, Kachura MA, Scallan CD, Geleziunas R, SenGupta D, Jooss K. Differential shaping of T cell responses elicited by heterologous ChAd68/self-amplifying mRNA SIV vaccine in macaques in combination with αCTLA4, αPD-1, or FLT3R agonist. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae052. [PMID: 40073084 DOI: 10.1093/jimmun/vkae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/03/2024] [Indexed: 03/14/2025]
Abstract
While therapeutic vaccines are a promising strategy for inducing human immunodeficiency virus (HIV) control, HIV vaccines tested to date have offered limited benefit to people living with HIV. The barriers to success may include the use of vaccine platforms and/or immunogens that drive weak or suboptimal immune responses, immune escape and/or immune dysfunction associated with chronic infection despite effective antiretroviral therapy. Combining vaccines with immune modulators in a safe manner may address some of the challenges and thus increase the efficacy of therapeutic HIV vaccines. We evaluated the immunogenicity of a ChAd68/samRNA-based simian immunodeficiency virus (SIV) vaccine regimen alone and in combination with a series of immune modulators in a preclinical rhesus macaque (M. mulatta) model. The vaccine was co-delivered with the checkpoint inhibitors αPD-1 or αCTLA-4, or with a FLT3 receptor agonist (FLT3Ra) shown to differentiate and expand dendritic cells and improve T cell priming. We demonstrate that the magnitude, breadth and functionality of SIV-specific vaccine-elicited CD8+ T cell responses were enhanced by combination with either αPD-1, αCTLA-4, or FLT3Ra. Combination with FLT3Ra also expanded polyfunctional CD4+ T cell responses. Our data demonstrate enhanced and distinct shaping of vaccine-elicited immune responses by immune modulators with implications for developing a functional HIV cure.
Collapse
Affiliation(s)
| | | | | | - Janette Sung
- Gilead Sciences, Inc, Foster City, CA, United States
| | - Brian Carr
- Gilead Sciences, Inc, Foster City, CA, United States
| | | | | | | | | | | | - Devi SenGupta
- Gilead Sciences, Inc, Foster City, CA, United States
| | - Karin Jooss
- Gritstone Bio, Inc, Emeryville, CA, United States
| |
Collapse
|
4
|
Al-Osaimi HM, Kanan M, Marghlani L, Al-Rowaili B, Albalawi R, Saad A, Alasmari S, Althobaiti K, Alhulaili Z, Alanzi A, Alqarni R, Alsofiyani R, Shrwani R. A systematic review on malaria and dengue vaccines for the effective management of these mosquito borne diseases: Improving public health. Hum Vaccin Immunother 2024; 20:2337985. [PMID: 38602074 PMCID: PMC11017952 DOI: 10.1080/21645515.2024.2337985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
Insect vector-borne diseases (VBDs) pose significant global health challenges, particularly in tropical and subtropical regions. The WHO has launched the "Global Vector Control Response (GVCR) 2017-2030" to address these diseases, emphasizing a comprehensive approach to vector control. This systematic review investigates the potential of malaria and dengue vaccines in controlling mosquito-borne VBDs, aiming to alleviate disease burdens and enhance public health. Following PRISMA 2020 guidelines, the review incorporated 39 new studies out of 934 identified records. It encompasses various studies assessing malaria and dengue vaccines, emphasizing the significance of vaccination as a preventive measure. The findings indicate variations in vaccine efficacy, duration of protection, and safety considerations for each disease, influencing public health strategies. The review underscores the urgent need for vaccines to combat the increasing burden of VBDs like malaria and dengue, advocating for ongoing research and investment in vaccine development.
Collapse
Affiliation(s)
- Hind M. Al-Osaimi
- Department of Pharmacy Services Administration, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh, Kingdom of Saudi Arabia
| | - Mohammed Kanan
- Department of Clinical Pharmacy, King Fahad Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Lujain Marghlani
- Department of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Badria Al-Rowaili
- Pharmaceutical Services Department, Northern Area Armed Forces Hospital, King Khalid Military, Hafr Al Batin, Kingdom of Saudi Arabia
| | - Reem Albalawi
- Department of Medicine, Tabuk University, Tabuk, Kingdom of Saudi Arabia
| | - Abrar Saad
- Pharmacy Department, Royal Commission Hospital, Yanbu, Kingdom of Saudi Arabia
| | - Saba Alasmari
- Department of Clinical Pharmacy, King Khalid University, Jeddah, Kingdom of Saudi Arabia
| | - Khaled Althobaiti
- Department of Medicine, Taif University, Ta’if, Kingdom of Saudi Arabia
| | - Zainab Alhulaili
- Department of Clinical Pharmacy, Dammam Medical Complex, Dammam, Kingdom of Saudi Arabia
| | - Abeer Alanzi
- Department of Medicine, King Abdulaziz Hospital, Makkah, Kingdom of Saudi Arabia
| | - Rawan Alqarni
- Department of Medicine and Surgery, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Razan Alsofiyani
- Department of Medicine, Taif University, Ta’if, Kingdom of Saudi Arabia
| | - Reem Shrwani
- Department of Clinical Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
5
|
Dobbs KR, Atieli HE, Valim C, Beeson JG. Previous Malaria Exposures and Immune Dysregulation: Developing Strategies To Improve Malaria Vaccine Efficacy in Young Children. Am J Trop Med Hyg 2024; 110:627-630. [PMID: 38442424 PMCID: PMC10993830 DOI: 10.4269/ajtmh.23-0696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/06/2023] [Indexed: 03/07/2024] Open
Abstract
After several decades in development, two malaria vaccines based on the same antigen and with very similar constructs and adjuvants, RTS,S/AS01 (RTS,S) and R21/Matrix-M (R21), were recommended by the WHO for widespread vaccination of children. These vaccines are much-needed additions to malaria control programs that, when used in conjunction with other control measures, will help to accelerate reductions in malaria morbidity and mortality. Although R21 is not yet available, RTS,S is currently being integrated into routine vaccine schedules in some areas. However, the efficacy of RTS,S is partial, short-lived, and varies widely according to age and geographic location. It is not clear why RTS,S induces protection in some individuals and not others, what the immune mechanisms are that favor protective immunity with RTS,S, and how immune mechanisms are influenced by host and environmental factors. Several studies suggest that higher levels of previous malaria exposure negatively impact RTS,S clinical efficacy. In this article, we summarize data suggesting that previous malaria exposures negatively impact the efficacy of RTS,S and other malaria vaccine candidates. We highlight recent evidence suggesting that increasing malaria exposure impairs the generation of functional antibody responses to RTS,S. Finally, we discuss how investigation of clinical and immune factors associated with suboptimal responses to RTS,S can be used to develop strategies to optimize RTS,S, which will remain relevant to R21 and next-generation vaccines.
Collapse
Affiliation(s)
| | | | - Clarissa Valim
- Boston University School of Public Health, Boston, Massachusetts
| | | |
Collapse
|
6
|
Rappaport AR, Kyi C, Lane M, Hart MG, Johnson ML, Henick BS, Liao CY, Mahipal A, Shergill A, Spira AI, Goldman JW, Scallan CD, Schenk D, Palmer CD, Davis MJ, Kounlavouth S, Kemp L, Yang A, Li YJ, Likes M, Shen A, Boucher GR, Egorova M, Veres RL, Espinosa JA, Jaroslavsky JR, Kraemer Tardif LD, Acrebuche L, Puccia C, Sousa L, Zhou R, Bae K, Hecht JR, Carbone DP, Johnson B, Allen A, Ferguson AR, Jooss K. A shared neoantigen vaccine combined with immune checkpoint blockade for advanced metastatic solid tumors: phase 1 trial interim results. Nat Med 2024; 30:1013-1022. [PMID: 38538867 DOI: 10.1038/s41591-024-02851-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/29/2024] [Indexed: 04/21/2024]
Abstract
Therapeutic vaccines that elicit cytotoxic T cell responses targeting tumor-specific neoantigens hold promise for providing long-term clinical benefit to patients with cancer. Here we evaluated safety and tolerability of a therapeutic vaccine encoding 20 shared neoantigens derived from selected common oncogenic driver mutations as primary endpoints in an ongoing phase 1/2 study in patients with advanced/metastatic solid tumors. Secondary endpoints included immunogenicity, overall response rate, progression-free survival and overall survival. Eligible patients were selected if their tumors expressed one of the human leukocyte antigen-matched tumor mutations included in the vaccine, with the majority of patients (18/19) harboring a mutation in KRAS. The vaccine regimen, consisting of a chimp adenovirus (ChAd68) and self-amplifying mRNA (samRNA) in combination with the immune checkpoint inhibitors ipilimumab and nivolumab, was shown to be well tolerated, with observed treatment-related adverse events consistent with acute inflammation expected with viral vector-based vaccines and immune checkpoint blockade, the majority grade 1/2. Two patients experienced grade 3/4 serious treatment-related adverse events that were also dose-limiting toxicities. The overall response rate was 0%, and median progression-free survival and overall survival were 1.9 months and 7.9 months, respectively. T cell responses were biased toward human leukocyte antigen-matched TP53 neoantigens encoded in the vaccine relative to KRAS neoantigens expressed by the patients' tumors, indicating a previously unknown hierarchy of neoantigen immunodominance that may impact the therapeutic efficacy of multiepitope shared neoantigen vaccines. These data led to the development of an optimized vaccine exclusively targeting KRAS-derived neoantigens that is being evaluated in a subset of patients in phase 2 of the clinical study. ClinicalTrials.gov registration: NCT03953235 .
Collapse
Affiliation(s)
| | - Chrisann Kyi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | - Brian S Henick
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Chih-Yi Liao
- University of Chicago Medical Center and Biological Sciences, Chicago, IL, USA
| | | | - Ardaman Shergill
- University of Chicago Medical Center and Biological Sciences, Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - David P Carbone
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | | | | | | | |
Collapse
|
7
|
Wajja A, Nassanga B, Natukunda A, Serubanja J, Tumusiime J, Akurut H, Oduru G, Nassuuna J, Kabagenyi J, Morrison H, Scott H, Doherty RP, Marshall JL, Puig IC, Cose S, Kaleebu P, Webb EL, Satti I, McShane H, Elliott AM. Safety and immunogenicity of ChAdOx1 85A prime followed by MVA85A boost compared with BCG revaccination among Ugandan adolescents who received BCG at birth: a randomised, open-label trial. THE LANCET. INFECTIOUS DISEASES 2024; 24:285-296. [PMID: 38012890 PMCID: PMC11876094 DOI: 10.1016/s1473-3099(23)00501-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND BCG confers reduced, variable protection against pulmonary tuberculosis. A more effective vaccine is needed. We evaluated the safety and immunogenicity of candidate regimen ChAdOx1 85A-MVA85A compared with BCG revaccination among Ugandan adolescents. METHODS After ChAdOx1 85A dose escalation and age de-escalation, we did a randomised open-label phase 2a trial among healthy adolescents aged 12-17 years, who were BCG vaccinated at birth, without evident tuberculosis exposure, in Entebbe, Uganda. Participants were randomly assigned (1:1) using a block size of 6, to ChAdOx1 85A followed by MVA85A (on day 56) or BCG (Moscow strain). Laboratory staff were masked to group assignment. Primary outcomes were solicited and unsolicited adverse events (AEs) up to day 28 and serious adverse events (SAEs) throughout the trial; and IFN-γ ELISpot response to antigen 85A (day 63 [geometric mean] and days 0-224 [area under the curve; AUC). FINDINGS Six adults (group 1, n=3; group 2, n=3) and six adolescents (group 3, n=3; group 4, n=3) were enrolled in the ChAdOx1 85A-only dose-escalation and age de-escalation studies (July to August, 2019). In the phase 2a trial, 60 adolescents were randomly assigned to ChAdOx1 85A-MVA85A (group 5, n=30) or BCG (group 6, n=30; December, 2019, to October, 2020). All 60 participants from groups 5 and 6 were included in the safety analysis, with 28 of 30 from group 5 (ChAdOx1 85A-MVA85A) and 29 of 30 from group 6 (BCG revaccination) analysed for immunogenicity outcomes. In the randomised trial, 60 AEs were reported among 23 (77%) of 30 participants following ChAdOx1 85A-MVA85A, 31 were systemic, with one severe event that occurred after the MVA85A boost that was rapidly self-limiting. All 30 participants in the BCG revaccination group reported at least one mild to moderate solicited AE; most were local reactions. There were no SAEs in either group. Ag85A-specific IFN-γ ELISpot responses peaked on day 63 in the ChAdOx1 85A-MVA85A group and were higher in the ChAdOx1 85A-MVA85A group compared with the BCG revaccination group (geometric mean ratio 30·59 [95% CI 17·46-53·59], p<0·0001, day 63; AUC mean difference 57 091 [95% CI 40 524-73 658], p<0·0001, days 0-224). INTERPRETATION The ChAdOx1 85A-MVA85A regimen was safe and induced stronger Ag85A-specific responses than BCG revaccination. Our findings support further development of booster tuberculosis vaccines. FUNDING UK Research and Innovations and Medical Research Council. TRANSLATIONS For the Swahili and Luganda translations of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Anne Wajja
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; Department of Global Health, Amsterdam University Medical Centers, Amsterdam, Netherlands; Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | - Beatrice Nassanga
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford, UK; Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda.
| | | | - Joel Serubanja
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | | | - Helen Akurut
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Gloria Oduru
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | | | | | - Hazel Morrison
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, University of Oxford, Churchill Hospital, Oxford, UK
| | - Hannah Scott
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | - Rebecca Powell Doherty
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | - Julia L Marshall
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | - Ingrid Cabrera Puig
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | - Stephen Cose
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | - Pontiano Kaleebu
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | - Emily L Webb
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Iman Satti
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | - Helen McShane
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford, UK; Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, University of Oxford, Churchill Hospital, Oxford, UK
| | - Alison M Elliott
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
8
|
Abstract
Malaria is a mosquito-borne disease caused by protozoan parasites of the genus Plasmodium. Despite significant declines in malaria-attributable morbidity and mortality over the last two decades, it remains a major public health burden in many countries. This underscores the critical need for improved strategies to prevent, treat and control malaria if we are to ultimately progress towards the eradication of this disease. Ideally, this will include the development and deployment of a highly effective malaria vaccine that is able to induce long-lasting protective immunity. There are many malaria vaccine candidates in development, with more than a dozen of these in clinical development. RTS,S/AS01 (also known as Mosquirix) is the most advanced malaria vaccine and was shown to have modest efficacy against clinical malaria in phase III trials in 5- to 17-month-old infants. Following pilot implementation trials, the World Health Organisation has recommended it for use in Africa in young children who are most at risk of infection with P. falciparum, the deadliest of the human malaria parasites. It is well recognised that more effective malaria vaccines are needed. In this review, we discuss malaria vaccine candidates that have progressed into clinical evaluation and highlight the most advanced candidates: Sanaria's irradiated sporozoite vaccine (PfSPZ Vaccine), the chemoattenuated sporozoite vaccine (PfSPZ-CVac), RTS,S/AS01 and the novel malaria vaccine candidate, R21, which displayed promising, high-level efficacy in a recent small phase IIb trial in Africa.
Collapse
Affiliation(s)
- Danielle I Stanisic
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| | - Michael F Good
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| |
Collapse
|
9
|
Silk SE, Kalinga WF, Mtaka IM, Lilolime NS, Mpina M, Milando F, Ahmed S, Diouf A, Mkwepu F, Simon B, Athumani T, Rashid M, Mohammed L, Lweno O, Ali AM, Nyaulingo G, Mwalimu B, Mswata S, Mwamlima TG, Barrett JR, Wang LT, Themistocleous Y, King LDW, Hodgson SH, Payne RO, Nielsen CM, Lawrie AM, Nugent FL, Cho JS, Long CA, Miura K, Draper SJ, Minassian AM, Olotu AI. Superior antibody immunogenicity of a viral-vectored RH5 blood-stage malaria vaccine in Tanzanian infants as compared to adults. MED 2023; 4:668-686.e7. [PMID: 37572659 DOI: 10.1016/j.medj.2023.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/23/2023] [Accepted: 07/11/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND RH5 is a leading blood-stage candidate antigen for a Plasmodium falciparum vaccine; however, its safety and immunogenicity in malaria-endemic populations are unknown. METHODS A phase 1b, single-center, dose-escalation, age-de-escalation, double-blind, randomized, controlled trial was conducted in Bagamoyo, Tanzania (NCT03435874). Between 12th April and 25th October 2018, 63 healthy adults (18-35 years), young children (1-6 years), and infants (6-11 months) received a priming dose of viral-vectored ChAd63 RH5 or rabies control vaccine. Sixty participants were boosted with modified vaccinia virus Ankara (MVA) RH5 or rabies control vaccine 8 weeks later and completed 6 months of follow-up post priming. Primary outcomes were the number of solicited and unsolicited adverse events post vaccination and the number of serious adverse events over the study period. Secondary outcomes included measures of the anti-RH5 immune response. FINDINGS Vaccinations were well tolerated, with profiles comparable across groups. No serious adverse events were reported. Vaccination induced RH5-specific cellular and humoral responses. Higher anti-RH5 serum immunoglobulin G (IgG) responses were observed post boost in young children and infants compared to adults. Vaccine-induced antibodies showed growth inhibition activity (GIA) in vitro against P. falciparum blood-stage parasites; their highest levels were observed in infants. CONCLUSIONS The ChAd63-MVA RH5 vaccine shows acceptable safety and reactogenicity and encouraging immunogenicity in children and infants residing in a malaria-endemic area. The levels of functional GIA observed in RH5-vaccinated infants are the highest reported to date following human vaccination. These data support onward clinical development of RH5-based blood-stage vaccines to protect against clinical malaria in young African infants. FUNDING Medical Research Council, London, UK.
Collapse
Affiliation(s)
- Sarah E Silk
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Wilmina F Kalinga
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Ivanny M Mtaka
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Nasoro S Lilolime
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Maximillian Mpina
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Florence Milando
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Saumu Ahmed
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Fatuma Mkwepu
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Beatus Simon
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Thabit Athumani
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Mohammed Rashid
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Latipha Mohammed
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Omary Lweno
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Ali M Ali
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Gloria Nyaulingo
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Bakari Mwalimu
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Sarah Mswata
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Tunu G Mwamlima
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| | - Jordan R Barrett
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Lawrence T Wang
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Yrene Themistocleous
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Lloyd D W King
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Susanne H Hodgson
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Ruth O Payne
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Carolyn M Nielsen
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Alison M Lawrie
- Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Fay L Nugent
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK
| | - Jee-Sun Cho
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK
| | - Carole A Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, MD 20852, USA
| | - Simon J Draper
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Angela M Minassian
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; Centre for Clinical Vaccinology and Tropical Medicine, Jenner Institute, University of Oxford, Old Road Campus, Oxford OX3 7LE, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford OX1 3QU, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Ally I Olotu
- Interventions and Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania
| |
Collapse
|
10
|
Tsoumani ME, Voyiatzaki C, Efstathiou A. Malaria Vaccines: From the Past towards the mRNA Vaccine Era. Vaccines (Basel) 2023; 11:1452. [PMID: 37766129 PMCID: PMC10536368 DOI: 10.3390/vaccines11091452] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Plasmodium spp. is the etiological agent of malaria, a life-threatening parasitic disease transmitted by infected mosquitoes. Malaria remains a major global health challenge, particularly in endemic regions. Over the years, various vaccine candidates targeting different stages of Plasmodium parasite life-cycle have been explored, including subunit vaccines, vectored vaccines, and whole organism vaccines with Mosquirix, a vaccine based on a recombinant protein, as the only currently approved vaccine for Plasmodium falciparum malaria. Despite the aforementioned notable progress, challenges such as antigenic diversity, limited efficacy, resistant parasites escaping protective immunity and the need for multiple doses have hindered the development of a highly efficacious malaria vaccine. The recent success of mRNA-based vaccines against SARS-CoV-2 has sparked renewed interest in mRNA vaccine platforms. The unique mRNA vaccine features, including their potential for rapid development, scalability, and flexibility in antigen design, make them a promising avenue for malaria vaccine development. This review provides an overview of the malaria vaccines' evolution from the past towards the mRNA vaccine era and highlights their advantages in overcoming the limitations of previous malaria vaccine candidates.
Collapse
Affiliation(s)
- Maria E. Tsoumani
- Department of Biomedical Sciences, University of West Attica, 12243 Aigaleo, Greece; (M.E.T.); (C.V.)
| | - Chrysa Voyiatzaki
- Department of Biomedical Sciences, University of West Attica, 12243 Aigaleo, Greece; (M.E.T.); (C.V.)
| | - Antonia Efstathiou
- Department of Biomedical Sciences, University of West Attica, 12243 Aigaleo, Greece; (M.E.T.); (C.V.)
- Immunology of Infection Group, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| |
Collapse
|
11
|
Nourani L, Mehrizi AA, Pirahmadi S, Pourhashem Z, Asadollahi E, Jahangiri B. CRISPR/Cas advancements for genome editing, diagnosis, therapeutics, and vaccine development for Plasmodium parasites, and genetic engineering of Anopheles mosquito vector. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 109:105419. [PMID: 36842543 DOI: 10.1016/j.meegid.2023.105419] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/30/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
Malaria as vector-borne disease remains important health concern with over 200 million cases globally. Novel antimalarial medicines and more effective vaccines must be developed to eliminate and eradicate malaria. Appraisal of preceding genome editing approaches confirmed the CRISPR/Cas nuclease system as a novel proficient genome editing system and a tool for species-specific diagnosis, and drug resistance researches for Plasmodium species, and gene drive to control Anopheles population. CRISPR/Cas technology, as a handy tool for genome editing can be justified for the production of transgenic malaria parasites like Plasmodium transgenic lines expressing Cas9, chimeric Plasmodium transgenic lines, knockdown and knockout transgenic parasites, and transgenic parasites expressing alternative alleles, and also mutant strains of Anopheles such as only male mosquito populations, generation of wingless mosquitoes, and creation of knock-out/ knock-in mutants. Though, the incorporation of traditional methods and novel molecular techniques could noticeably enhance the quality of results. The striking development of a CRISPR/Cas-based diagnostic kit that can specifically diagnose the Plasmodium species or drug resistance markers is highly required in malaria settings with affordable cost and high-speed detection. Furthermore, the advancement of genome modifications by CRISPR/Cas technologies resolves contemporary restrictions to culturing, maintaining, and analyzing these parasites, and the aptitude to investigate parasite genome functions opens up new vistas in the better understanding of pathogenesis.
Collapse
Affiliation(s)
- Leila Nourani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Zeinab Pourhashem
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Elahe Asadollahi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Babak Jahangiri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
12
|
Recent Advances in the Development of Adenovirus-Vectored Vaccines for Parasitic Infections. Pharmaceuticals (Basel) 2023; 16:ph16030334. [PMID: 36986434 PMCID: PMC10058461 DOI: 10.3390/ph16030334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Vaccines against parasites have lagged centuries behind those against viral and bacterial infections, despite the devastating morbidity and widespread effects of parasitic diseases across the globe. One of the greatest hurdles to parasite vaccine development has been the lack of vaccine strategies able to elicit the complex and multifaceted immune responses needed to abrogate parasitic persistence. Viral vectors, especially adenovirus (AdV) vectors, have emerged as a potential solution for complex disease targets, including HIV, tuberculosis, and parasitic diseases, to name a few. AdVs are highly immunogenic and are uniquely able to drive CD8+ T cell responses, which are known to be correlates of immunity in infections with most protozoan and some helminthic parasites. This review presents recent developments in AdV-vectored vaccines targeting five major human parasitic diseases: malaria, Chagas disease, schistosomiasis, leishmaniasis, and toxoplasmosis. Many AdV-vectored vaccines have been developed for these diseases, utilizing a wide variety of vectors, antigens, and modes of delivery. AdV-vectored vaccines are a promising approach for the historically challenging target of human parasitic diseases.
Collapse
|
13
|
Malaria Vaccines. Infect Dis (Lond) 2023. [DOI: 10.1007/978-1-0716-2463-0_536] [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: 02/10/2023] Open
|
14
|
Morter R, Tiono AB, Nébié I, Hague O, Ouedraogo A, Diarra A, Viebig NK, Hill AVS, Ewer KJ, Sirima SB. Impact of exposure to malaria and nutritional status on responses to the experimental malaria vaccine ChAd63 MVA ME-TRAP in 5-17 month-old children in Burkina Faso. Front Immunol 2022; 13:1058227. [PMID: 36532031 PMCID: PMC9755991 DOI: 10.3389/fimmu.2022.1058227] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
The experimental malaria vaccine ChAd63 MVA ME-TRAP previously showed protective efficacy against Plasmodium falciparum infection in Phase IIa sporozoite challenge studies in adults in the United Kingdom and in a Phase IIb field efficacy trial in Kenyan adults. However, it failed to demonstrate efficacy in a phase IIb trial in 5-17 month-old children in an area of high malaria transmission in Burkina Faso. This secondary analysis investigated whether exposure to malaria or nutritional status might be associated with reduced responses to vaccination in this cohort. Parasite blood smears and anti-AMA-1 IgG titres were used to assess history of exposure to malaria and weight-for-length Z scores were calculated to assess nutritional status. Differences in vaccine-specific anti-TRAP IgG titre and ex vivo IFNγ ELISpot response were measured between groups. In total, n = 336 volunteers randomised to receive the experimental vaccine regimen were included in this analysis. A positive smear microscopy result was associated with reduced anti-TRAP IgG titre (geometric mean titre: 2775 (uninfected) vs 1968 (infected), p = 0.025), whilst anti-AMA-1 IgG titres were weakly negatively correlated with reduced ex vivo IFNγ ELISpot response (r = -0.18, p = 0.008). Nutritional status was not associated with either humoral or cellular immunogenicity. Vaccine efficacy was also measured separately for vaccinees with positive and negative blood smears. Although not significant in either group compared to controls, vaccine efficacy measured by Cox hazard ratio was higher in uninfected compared to infected individuals (19.8% [p = 0.50] vs 3.3% [p = 0.69]). Overall, this data suggests exposure to malaria may be associated with impaired vaccine immunogenicity. This may have consequences for the testing and eventual deployment of various vaccines, in areas with high endemicity for malaria. Trial Registration Pactr.org, identifier PACTR201208000404131; ClinicalTrials.gov, identifier NCT01635647.
Collapse
Affiliation(s)
- Richard Morter
- Nuffield Department of Clinical Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alfred B. Tiono
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso,Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Issa Nébié
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Oliver Hague
- Nuffield Department of Clinical Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Amidou Diarra
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Nicola K. Viebig
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Adrian V. S. Hill
- Nuffield Department of Clinical Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Katie J. Ewer
- Nuffield Department of Clinical Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom,*Correspondence: Sodiomon B. Sirima, ; Katie J. Ewer,
| | - Sodiomon B. Sirima
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso,*Correspondence: Sodiomon B. Sirima, ; Katie J. Ewer,
| |
Collapse
|
15
|
Pischel L, Patel KM, Goshua G, Omer SB. Adenovirus-Based Vaccines and Thrombosis in Pregnancy: A Systematic Review and Meta-analysis. Clin Infect Dis 2022; 75:1179-1186. [PMID: 35134164 PMCID: PMC9383370 DOI: 10.1093/cid/ciac080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Rare cases of thrombosis and thrombocytopenia (thrombosis with thrombocytopenia syndrome [TTS]) have been associated with 2 coronavirus disease 2019 adenovirus vector vaccines: the ChAdOx1 nCoV-19 Vaxzevria vaccine (Oxford/AstraZeneca) and the JNJ-7836735 Johnson & Johnson vaccine (Janssen). It is unknown if TTS is a class-mediated effect of adenovirus-based vaccines or if it could worsen known hypercoagulable states. Since most cases of TTS happen in women of childbearing age, pregnancy is a crucial risk factor to assess. Understanding these risks is important for advising vaccine recipients and future adenovirus vector vaccine development. METHODS To explore the potential associations of adenovirus-based vaccine components with symptoms of TTS in the general clinical trial population and in pregnant women in clinical trials, we conducted a systematic review and meta-analysis of adenovirus-based vector vaccines to document cases of thrombocytopenia, coagulopathy, and or pregnancy from 1 January 1966 to 9 August 2021. RESULTS We found 167 articles from 159 studies of adenovirus vector-based vaccines, 123 of which targeted infectious diseases. In the general population, 20 studies reported an event of thrombocytopenia and 20 studies indicated some coagulopathy. Among pregnant women, of the 28 studies that reported a total of 1731 pregnant women, thrombocytopenia or coagulopathy were not reported. CONCLUSIONS In this systematic review and meta-analysis, there was no class-wide effect of adenovirus vector vaccines toward thrombocytopenia or coagulopathy events in the general population or in pregnant women.
Collapse
Affiliation(s)
- Lauren Pischel
- Correspondence: L. Pischel, Section of Infectious Diseases, Yale School of Medicine, 135 College St, Suite 323, New Haven, CT 06510-2483 ()
| | - Kavin M Patel
- Section of Infectious Diseases, Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - George Goshua
- Section of Hematology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Health Policy and Management, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Saad B Omer
- Section of Infectious Diseases, Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Yale School of Public Health, New Haven, Connecticut, USA
- Yale Institute of Global Health, New Haven, Connecticut, USA
- Yale School of Nursing, Orange, Connecticut, USA
| |
Collapse
|
16
|
Owalla TJ, Hergott DEB, Seilie AM, Staubus W, Chavtur C, Chang M, Kublin JG, Egwang TG, Murphy SC. Rethinking detection of pre-existing and intervening Plasmodium infections in malaria clinical trials. Front Immunol 2022; 13:1003452. [PMID: 36203582 PMCID: PMC9531235 DOI: 10.3389/fimmu.2022.1003452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/22/2022] [Indexed: 02/02/2023] Open
Abstract
Pre-existing and intervening low-density Plasmodium infections complicate the conduct of malaria clinical trials. These infections confound infection detection endpoints, and their immunological effects may detract from intended vaccine-induced immune responses. Historically, these infections were often unrecognized since infrequent and often analytically insensitive parasitological testing was performed before and during trials. Molecular diagnostics now permits their detection, but investigators must weigh the cost, complexity, and personnel demands on the study and the laboratory when scheduling such tests. This paper discusses the effect of pre-existing and intervening, low-density Plasmodium infections on malaria vaccine trial endpoints and the current methods employed for their infection detection. We review detection techniques, that until recently, provided a dearth of cost-effective strategies for detecting low density infections. A recently deployed, field-tested, simple, and cost-effective molecular diagnostic strategy for detecting pre-existing and intervening Plasmodium infections from dried blood spots (DBS) in malaria-endemic settings is discussed to inform new clinical trial designs. Strategies that combine sensitive molecular diagnostic techniques with convenient DBS collections and cost-effective pooling strategies may enable more thorough and informative infection monitoring in upcoming malaria clinical trials and epidemiological studies.
Collapse
Affiliation(s)
- Tonny J. Owalla
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Dianna E. B. Hergott
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
| | - Annette M. Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Weston Staubus
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Chris Chavtur
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Ming Chang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States
| | - James G. Kublin
- Department of Global Health, University of Washington, Seattle, WA, United States,Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Thomas G. Egwang
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States,Seattle Malaria Clinical Trials Center, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States,Department of Microbiology, University of Washington, Seattle, WA, United States,*Correspondence: Sean C. Murphy,
| |
Collapse
|
17
|
Nunes-Cabaço H, Moita D, Prudêncio M. Five decades of clinical assessment of whole-sporozoite malaria vaccines. Front Immunol 2022; 13:977472. [PMID: 36159849 PMCID: PMC9493004 DOI: 10.3389/fimmu.2022.977472] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
In 1967, pioneering work by Ruth Nussenzweig demonstrated for the first time that irradiated sporozoites of the rodent malaria parasite Plasmodium berghei protected mice against a challenge with infectious parasites of the same species. This remarkable finding opened up entirely new prospects of effective vaccination against malaria using attenuated sporozoites as immunization agents. The potential for whole-sporozoite-based immunization in humans was established in a clinical study in 1973, when a volunteer exposed to X-irradiated P. falciparum sporozoites was found to be protected against malaria following challenge with a homologous strain of this parasite. Nearly five decades later, much has been achieved in the field of whole-sporozoite malaria vaccination, and multiple reports on the clinical evaluation of such candidates have emerged. However, this process has known different paces before and after the turn of the century. While only a few clinical studies were published in the 1970’s, 1980’s and 1990’s, remarkable progress was made in the 2000’s and beyond. This article reviews the history of the clinical assessment of whole-sporozoite malaria vaccines over the last forty-nine years, highlighting the impressive achievements made over the last few years, and discussing some of the challenges ahead.
Collapse
|
18
|
Individualized, heterologous chimpanzee adenovirus and self-amplifying mRNA neoantigen vaccine for advanced metastatic solid tumors: phase 1 trial interim results. Nat Med 2022; 28:1619-1629. [PMID: 35970920 DOI: 10.1038/s41591-022-01937-6] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/06/2022] [Indexed: 12/30/2022]
Abstract
Checkpoint inhibitor (CPI) therapies provide limited benefit to patients with tumors of low immune reactivity. T cell-inducing vaccines hold promise to exert long-lasting disease control in combination with CPI therapy. Safety, tolerability and recommended phase 2 dose (RP2D) of an individualized, heterologous chimpanzee adenovirus (ChAd68) and self-amplifying mRNA (samRNA)-based neoantigen vaccine in combination with nivolumab and ipilimumab were assessed as primary endpoints in an ongoing phase 1/2 study in patients with advanced metastatic solid tumors (NCT03639714). The individualized vaccine regimen was safe and well tolerated, with no dose-limiting toxicities. Treatment-related adverse events (TRAEs) >10% included pyrexia, fatigue, musculoskeletal and injection site pain and diarrhea. Serious TRAEs included one count each of pyrexia, duodenitis, increased transaminases and hyperthyroidism. The RP2D was 1012 viral particles (VP) ChAd68 and 30 µg samRNA. Secondary endpoints included immunogenicity, feasibility of manufacturing and overall survival (OS). Vaccine manufacturing was feasible, with vaccination inducing long-lasting neoantigen-specific CD8 T cell responses. Several patients with microsatellite-stable colorectal cancer (MSS-CRC) had improved OS. Exploratory biomarker analyses showed decreased circulating tumor DNA (ctDNA) in patients with prolonged OS. Although small study size limits statistical and translational analyses, the increased OS observed in MSS-CRC warrants further exploration in larger randomized studies.
Collapse
|
19
|
Wilder BK, Vigdorovich V, Carbonetti S, Minkah N, Hertoghs N, Raappana A, Cardamone H, Oliver BG, Trakhimets O, Kumar S, Dambrauskas N, Arredondo SA, Camargo N, Seilie AM, Murphy SC, Kappe SHI, Sather DN. Anti-TRAP/SSP2 monoclonal antibodies can inhibit sporozoite infection and may enhance protection of anti-CSP monoclonal antibodies. NPJ Vaccines 2022; 7:58. [PMID: 35618791 PMCID: PMC9135708 DOI: 10.1038/s41541-022-00480-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 04/22/2022] [Indexed: 11/10/2022] Open
Abstract
Vaccine-induced sterilizing protection from infection by Plasmodium parasites, the pathogens that cause malaria, will be essential in the fight against malaria as it would prevent both malaria-related disease and transmission. Stopping the relatively small number of parasites injected by the mosquito before they can migrate from the skin to the liver is an attractive means to this goal. Antibody-eliciting vaccines have been used to pursue this objective by targeting the major parasite surface protein present during this stage, the circumsporozoite protein (CSP). While CSP-based vaccines have recently had encouraging success in disease reduction, this was only achieved with extremely high antibody titers and appeared less effective for a complete block of infection (i.e., sterile protection). While such disease reduction is important, these and other results indicate that strategies focusing on CSP alone may not achieve the high levels of sterile protection needed for malaria eradication. Here, we show that monoclonal antibodies (mAbs) recognizing another sporozoite protein, TRAP/SSP2, exhibit a range of inhibitory activity and that these mAbs may augment CSP-based protection despite conferring no sterile protection on their own. Therefore, pursuing a multivalent subunit vaccine immunization is a promising strategy for improving infection-blocking malaria vaccines.
Collapse
Affiliation(s)
- Brandon K Wilder
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Vladimir Vigdorovich
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sara Carbonetti
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nana Minkah
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nina Hertoghs
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Andrew Raappana
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Hayley Cardamone
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Brian G Oliver
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Olesya Trakhimets
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nicholas Dambrauskas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Silvia A Arredondo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Annette M Seilie
- Department of Laboratory Medicine and Pathology and Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology and Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
20
|
Noé A, Datoo MS, Flaxman A, Husainy MA, Jenkin D, Bellamy D, Makinson RA, Morter R, Ramos Lopez F, Sheridan J, Voukantsis D, Prasad N, Hill AVS, Ewer KJ, Spencer AJ. Deep Immune Phenotyping and Single-Cell Transcriptomics Allow Identification of Circulating TRM-Like Cells Which Correlate With Liver-Stage Immunity and Vaccine-Induced Protection From Malaria. Front Immunol 2022; 13:795463. [PMID: 35197971 PMCID: PMC8859435 DOI: 10.3389/fimmu.2022.795463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
Protection from liver-stage malaria requires high numbers of CD8+ T cells to find and kill Plasmodium-infected cells. A new malaria vaccine strategy, prime-target vaccination, involves sequential viral-vectored vaccination by intramuscular and intravenous routes to target cellular immunity to the liver. Liver tissue-resident memory (TRM) CD8+ T cells have been shown to be necessary and sufficient for protection against rodent malaria by this vaccine regimen. Ultimately, to most faithfully assess immunotherapeutic responses by these local, specialised, hepatic T cells, periodic liver sampling is necessary, however this is not feasible at large scales in human trials. Here, as part of a phase I/II P. falciparum challenge study of prime-target vaccination, we performed deep immune phenotyping, single-cell RNA-sequencing and kinetics of hepatic fine needle aspirates and peripheral blood samples to study liver CD8+ TRM cells and circulating counterparts. We found that while these peripheral ‘TRM-like’ cells differed to TRM cells in terms of previously described characteristics, they are similar phenotypically and indistinguishable in terms of key T cell residency transcriptional signatures. By exploring the heterogeneity among liver CD8+ TRM cells at single cell resolution we found two main subpopulations that each share expression profiles with blood T cells. Lastly, our work points towards the potential for using TRM−like cells as a correlate of protection by liver-stage malaria vaccines and, in particular, those adopting a prime-target approach. A simple and reproducible correlate of protection would be particularly valuable in trials of liver-stage malaria vaccines as they progress to phase III, large-scale testing in African infants. We provide a blueprint for understanding and monitoring liver TRM cells induced by a prime-target malaria vaccine approach.
Collapse
Affiliation(s)
- Andrés Noé
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- *Correspondence: Andrés Noé, ; ; Alexandra J. Spencer,
| | - Mehreen S. Datoo
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Amy Flaxman
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Daniel Jenkin
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Duncan Bellamy
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Richard Morter
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Dimitrios Voukantsis
- Bioinformatics Hub, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Naveen Prasad
- Bioinformatics Hub, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Katie J. Ewer
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alexandra J. Spencer
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- *Correspondence: Andrés Noé, ; ; Alexandra J. Spencer,
| |
Collapse
|
21
|
Lefebvre MN, Drewry LL, Pewe LL, Hancox LS, Reyes-Sandoval A, Harty JT. Cutting Edge: Subunit Booster Vaccination Confers Sterilizing Immunity against Liver-Stage Malaria in Mice Initially Primed with a Weight-Normalized Dose of Radiation-Attenuated Sporozoites. THE JOURNAL OF IMMUNOLOGY 2021; 207:2631-2635. [PMID: 34716185 DOI: 10.4049/jimmunol.2100818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/25/2021] [Indexed: 11/19/2022]
Abstract
Radiation-attenuated sporozoite (RAS) vaccination offers hope for global malaria control through induction of protective liver-stage-specific memory CD8 T cells. Effective RAS vaccination regimens exist; however, widespread implementation remains unfeasible. A key difficulty resides in the need to administer three or more doses i.v. to achieve sufficient immunity. Strategies to reduce the number of RAS doses are therefore desirable. Here we used mice to model human immune responses to a single, suboptimal weight-normalized RAS dose administered i.v. followed by subunit vaccination to amplify liver-stage-specific memory CD8 T cells. RAS+subunit prime-boost regimens increased the numbers of liver-stage-specific memory CD8 T cells to a level greater than is present after one RAS vaccination. Both i.v. and i.m. subunit vaccine delivery induced immunity in mice, and many vaccinated mice completely cleared liver infection. These findings are particularly relevant to human vaccine development because RAS+subunit prime-boost vaccination would reduce the logistical challenges of multiple RAS-only immunizations.
Collapse
Affiliation(s)
- Mitchell N Lefebvre
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA; and
| | - Lisa L Drewry
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lecia L Pewe
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lisa S Hancox
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | - John T Harty
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; .,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA; and
| |
Collapse
|
22
|
Pirahmadi S, Afzali S, Zargar M, Zakeri S, Mehrizi AA. How can we develop an effective subunit vaccine to achieve successful malaria eradication? Microb Pathog 2021; 160:105203. [PMID: 34547408 DOI: 10.1016/j.micpath.2021.105203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/05/2021] [Accepted: 09/17/2021] [Indexed: 12/16/2022]
Abstract
Malaria, a mosquito-borne infection, is the most widespread parasitic disease. Despite numerous efforts to eradicate malaria, this disease is still a health concern worldwide. Owing to insecticide-resistant vectors and drug-resistant parasites, available controlling measures are insufficient to achieve a malaria-free world. Thus, there is an urgent need for new intervention tools such as efficient malaria vaccines. Subunit vaccines are the most promising malaria vaccines under development. However, one of the major drawbacks of subunit vaccines is the lack of efficient and durable immune responses including antigen-specific antibody, CD4+, and CD8+ T-cell responses, long-lived plasma cells, memory cells, and functional antibodies for parasite neutralization or inhibition of parasite invasion. These types of responses could be induced by whole organism vaccines, but eliciting these responses with subunit vaccines has been proven to be more challenging. Consequently, subunit vaccines require several policies to overcome these challenges. In this review, we address common approaches that can improve the efficacy of subunit vaccines against malaria.
Collapse
Affiliation(s)
- Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Shima Afzali
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Zargar
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
23
|
Gibbins MP, Müller K, Matuschewski K, Silvie O, Hafalla JCR. Inferior T cell immunogenicity of a Plasmodium berghei model liver stage antigen expressed throughout pre-erythrocytic maturation. Parasite Immunol 2021; 43:e12877. [PMID: 34515999 DOI: 10.1111/pim.12877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/01/2022]
Abstract
Sporozoite antigens are the basis of a number of malaria vaccines being tested, but the contribution of antigens expressed during subsequent liver stage development to pre-erythrocytic stage immunity is poorly understood. We previously showed that, following immunisation with radiation attenuated sporozoites (RAS), a model epitope embedded in a sporozoite surface protein elicited robust CD8+ T cell responses, whilst the same epitope in a liver stage antigen induced inferior responses. Since RAS arrest early in their development in host hepatocytes, we hypothesised that extending parasite maturation in the liver could considerably improve the epitope-specific CD8+ T cell response. Here, we employed a late liver stage arrested parasite model, azithromycin prophylaxis alongside live sporozoites, to increase expression of the model epitope until full liver stage maturation. Strikingly, this alternative immunisation strategy, which has been shown to elicit superior protection, failed to improve the resulting epitope-specific CD8+ T cell responses. Our findings support the notion that liver stage antigens are poorly immunogenic and provide additional caution about prioritising antigens for vaccine development based solely on immunogenicity.
Collapse
Affiliation(s)
- Matthew P Gibbins
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Katja Müller
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Institute of Biology, Humboldt University, Berlin, Germany
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Institute of Biology, Humboldt University, Berlin, Germany
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Julius Clemence R Hafalla
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
24
|
Kolli SK, Salman AM, Ramesar J, Chevalley-Maurel S, Kroeze H, Geurten FGA, Miyazaki S, Mukhopadhyay E, Marin-Mogollon C, Franke-Fayard B, Hill AVS, Janse CJ. Screening of viral-vectored P. falciparum pre-erythrocytic candidate vaccine antigens using chimeric rodent parasites. PLoS One 2021; 16:e0254498. [PMID: 34252120 PMCID: PMC8274855 DOI: 10.1371/journal.pone.0254498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/28/2021] [Indexed: 11/19/2022] Open
Abstract
To screen for additional vaccine candidate antigens of Plasmodium pre-erythrocytic stages, fourteen P. falciparum proteins were selected based on expression in sporozoites or their role in establishment of hepatocyte infection. For preclinical evaluation of immunogenicity of these proteins in mice, chimeric P. berghei sporozoites were created that express the P. falciparum proteins in sporozoites as an additional copy gene under control of the uis4 gene promoter. All fourteen chimeric parasites produced sporozoites but sporozoites of eight lines failed to establish a liver infection, indicating a negative impact of these P. falciparum proteins on sporozoite infectivity. Immunogenicity of the other six proteins (SPELD, ETRAMP10.3, SIAP2, SPATR, HT, RPL3) was analyzed by immunization of inbred BALB/c and outbred CD-1 mice with viral-vectored (ChAd63 or ChAdOx1, MVA) vaccines, followed by challenge with chimeric sporozoites. Protective immunogenicity was determined by analyzing parasite liver load and prepatent period of blood stage infection after challenge. Of the six proteins only SPELD immunized mice showed partial protection. We discuss both the low protective immunogenicity of these proteins in the chimeric rodent malaria challenge model and the negative effect on P. berghei sporozoite infectivity of several P. falciparum proteins expressed in the chimeric sporozoites.
Collapse
Affiliation(s)
- Surendra Kumar Kolli
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Ahmed M. Salman
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jai Ramesar
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Hans Kroeze
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Fiona G. A. Geurten
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Shinya Miyazaki
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Ekta Mukhopadhyay
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Adrian V. S. Hill
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Chris J. Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
25
|
Steel RWJ, Vigdorovich V, Dambrauskas N, Wilder BK, Arredondo SA, Goswami D, Kumar S, Carbonetti S, Swearingen KE, Nguyen T, Betz W, Camargo N, Fisher BS, Soden J, Thomas H, Freeth J, Moritz RL, Noah Sather D, Kappe SHI. Platelet derived growth factor receptor β (PDGFRβ) is a host receptor for the human malaria parasite adhesin TRAP. Sci Rep 2021; 11:11328. [PMID: 34059712 PMCID: PMC8166973 DOI: 10.1038/s41598-021-90722-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/13/2021] [Indexed: 02/04/2023] Open
Abstract
Following their inoculation by the bite of an infected Anopheles mosquito, the malaria parasite sporozoite forms travel from the bite site in the skin into the bloodstream, which transports them to the liver. The thrombospondin-related anonymous protein (TRAP) is a type 1 transmembrane protein that is released from secretory organelles and relocalized on the sporozoite plasma membrane. TRAP is required for sporozoite motility and host infection, and its extracellular portion contains adhesive domains that are predicted to engage host receptors. Here, we identified the human platelet-derived growth factor receptor β (hPDGFRβ) as one such protein receptor. Deletion constructs showed that the von Willebrand factor type A and thrombospondin repeat domains of TRAP are both required for optimal binding to hPDGFRβ-expressing cells. We also demonstrate that this interaction is conserved in the human-infective parasite Plasmodium vivax, but not the rodent-infective parasite Plasmodium yoelii. We observed expression of hPDGFRβ mainly in cells associated with the vasculature suggesting that TRAP:hPDGFRβ interaction may play a role in the recognition of blood vessels by invading sporozoites.
Collapse
Affiliation(s)
- Ryan W J Steel
- Seattle Children's Research Institute, Seattle, WA, USA
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | | | | | - Brandon K Wilder
- Seattle Children's Research Institute, Seattle, WA, USA
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | | | | | - Sudhir Kumar
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | - Thao Nguyen
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Will Betz
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Jo Soden
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | - Helen Thomas
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | - Jim Freeth
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | | | - D Noah Sather
- Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Stefan H I Kappe
- Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
26
|
Bezbaruah R, Borah P, Kakoti BB, Al-Shar’I NA, Chandrasekaran B, Jaradat DMM, Al-Zeer MA, Abu-Romman S. Developmental Landscape of Potential Vaccine Candidates Based on Viral Vector for Prophylaxis of COVID-19. Front Mol Biosci 2021; 8:635337. [PMID: 33937326 PMCID: PMC8082173 DOI: 10.3389/fmolb.2021.635337] [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: 11/30/2020] [Accepted: 02/05/2021] [Indexed: 12/18/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2, SARS-CoV-2, arose at the end of 2019 as a zoonotic virus, which is the causative agent of the novel coronavirus outbreak COVID-19. Without any clear indications of abatement, the disease has become a major healthcare threat across the globe, owing to prolonged incubation period, high prevalence, and absence of existing drugs or vaccines. Development of COVID-19 vaccine is being considered as the most efficient strategy to curtail the ongoing pandemic. Following publication of genetic sequence of SARS-CoV-2, globally extensive research and development work has been in progress to develop a vaccine against the disease. The use of genetic engineering, recombinant technologies, and other computational tools has led to the expansion of several promising vaccine candidates. The range of technology platforms being evaluated, including virus-like particles, peptides, nucleic acid (DNA and RNA), recombinant proteins, inactivated virus, live attenuated viruses, and viral vectors (replicating and non-replicating) approaches, are striking features of the vaccine development strategies. Viral vectors, the next-generation vaccine platforms, provide a convenient method for delivering vaccine antigens into the host cell to induce antigenic proteins which can be tailored to arouse an assortment of immune responses, as evident from the success of smallpox vaccine and Ervebo vaccine against Ebola virus. As per the World Health Organization, till January 22, 2021, 14 viral vector vaccine candidates are under clinical development including 10 nonreplicating and four replicating types. Moreover, another 39 candidates based on viral vector platform are under preclinical evaluation. This review will outline the current developmental landscape and discuss issues that remain critical to the success or failure of viral vector vaccine candidates against COVID-19.
Collapse
Affiliation(s)
- Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | - Pobitra Borah
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| | - Bibhuti Bhushan Kakoti
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | - Nizar A. Al-Shar’I
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Da’san M. M. Jaradat
- Department of Chemistry, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Munir A. Al-Zeer
- Department of Applied Biochemistry, Institute of Biotechnology, Technical University of Berlin, Berlin, Germany
| | - Saeid Abu-Romman
- Department of Biotechnology, Faculty of Agricultural Technology, Al-Balqa Applied University, Al-Salt, Jordan
| |
Collapse
|
27
|
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
|
28
|
Lu G, Shan S, Zainab B, Ayaz Z, He J, Xie Z, Rashid U, Zhang D, Mehmood Abbasi A. Novel vaccine design based on genomics data analysis: A review. Scand J Immunol 2021; 93:e12986. [PMID: 33043473 DOI: 10.1111/sji.12986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/28/2022]
Abstract
Modification of pathogenic strains with the passage of time is responsible for evolution in the timeline of vaccine development for last 30 years. Recent advancements in computational vaccinology on the one hand and genome sequencing approaches on the other have generated new hopes in vaccine development. The aim of this review was to discuss the evolution of vaccines, their characteristics and limitations. In this review, we highlighted the evolution of vaccines, from first generation to the current status, pointing out how different vaccines have emerged and different approaches that are being followed up in the development of more rational vaccines against a wide range of diseases. Data were collected using Google Scholar, Web of Science, Science Direct, Web of Knowledge, Scopus and Science Hub, whereas computational tools such as NCBI, GeneMANIA and STRING were used to analyse the pathways of vaccine action. Innovative tools, such as computational tools, recombinant technologies and intra-dermal devices, are currently being investigated in order to improve the immunological response. New technologies enlightened the interactions of host proteins with pathogenic proteins for vaccine candidate development, but still there is a need of integrating transcriptomic and proteomic approaches. Although immunization with genomics data is a successful approach, its advantages must be assessed case by case and its applicability depends on the nature of the agent to be immunized, the nature of the antigen and the type of immune response required to achieve effective protection.
Collapse
Affiliation(s)
- Guangli Lu
- Institute of Business, School of Business, Henan University, Henan, China
| | - Sharui Shan
- The First Affiliated Hospital of Jinan University (Guangzhou Overseas Chinese Hospital), Guangzhou, China
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Bibi Zainab
- Department of Environmental Sciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Zainab Ayaz
- Department of Environmental Sciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Jialiang He
- School of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Zhenxing Xie
- Basic School of Medicine, Henan University, Kaifeng, China
| | - Umer Rashid
- Department of Chemistry, COMSATS University Islamabad, Islamabad, Pakistan
| | - Dalin Zhang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Arshad Mehmood Abbasi
- Department of Environmental Sciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
29
|
Pance A. The Stem Cell Revolution Revealing Protozoan Parasites' Secrets and Paving the Way towards Vaccine Development. Vaccines (Basel) 2021; 9:105. [PMID: 33572549 PMCID: PMC7911700 DOI: 10.3390/vaccines9020105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Protozoan infections are leading causes of morbidity and mortality in humans and some of the most important neglected diseases in the world. Despite relentless efforts devoted to vaccine and drug development, adequate tools to treat and prevent most of these diseases are still lacking. One of the greatest hurdles is the lack of understanding of host-parasite interactions. This gap in our knowledge comes from the fact that these parasites have complex life cycles, during which they infect a variety of specific cell types that are difficult to access or model in vitro. Even in those cases when host cells are readily available, these are generally terminally differentiated and difficult or impossible to manipulate genetically, which prevents assessing the role of human factors in these diseases. The advent of stem cell technology has opened exciting new possibilities to advance our knowledge in this field. The capacity to culture Embryonic Stem Cells, derive Induced Pluripotent Stem Cells from people and the development of protocols for differentiation into an ever-increasing variety of cell types and organoids, together with advances in genome editing, represent a huge resource to finally crack the mysteries protozoan parasites hold and unveil novel targets for prevention and treatment.
Collapse
Affiliation(s)
- Alena Pance
- The Wellcome Sanger Institute, Genome Campus, Hinxton Cambridgeshire CB10 1SA, UK
| |
Collapse
|
30
|
Sasso E, D'Alise AM, Zambrano N, Scarselli E, Folgori A, Nicosia A. New viral vectors for infectious diseases and cancer. Semin Immunol 2020; 50:101430. [PMID: 33262065 DOI: 10.1016/j.smim.2020.101430] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/23/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Since the discovery in 1796 by Edward Jenner of vaccinia virus as a way to prevent and finally eradicate smallpox, the concept of using a virus to fight another virus has evolved into the current approaches of viral vectored genetic vaccines. In recent years, key improvements to the vaccinia virus leading to a safer version (Modified Vaccinia Ankara, MVA) and the discovery that some viruses can be used as carriers of heterologous genes encoding for pathological antigens of other infectious agents (the concept of 'viral vectors') has spurred a new wave of clinical research potentially providing for a solution for the long sought after vaccines against major diseases such as HIV, TB, RSV and Malaria, or emerging infectious diseases including those caused by filoviruses and coronaviruses. The unique ability of some of these viral vectors to stimulate the cellular arm of the immune response and, most importantly, T lymphocytes with cell killing activity, has also reawakened the interest toward developing therapeutic vaccines against chronic infectious diseases and cancer. To this end, existing vectors such as those based on Adenoviruses have been improved in immunogenicity and efficacy. Along the same line, new vectors that exploit viruses such as Vesicular Stomatitis Virus (VSV), Measles Virus (MV), Lymphocytic choriomeningitis virus (LCMV), cytomegalovirus (CMV), and Herpes Simplex Virus (HSV), have emerged. Furthermore, technological progress toward modifying their genome to render some of these vectors incompetent for replication has increased confidence toward their use in infant and elderly populations. Lastly, their production process being the same for every product has made viral vectored vaccines the technology of choice for rapid development of vaccines against emerging diseases and for 'personalised' cancer vaccines where there is an absolute need to reduce time to the patient from months to weeks or days. Here we review the recent developments in viral vector technologies, focusing on novel vectors based on primate derived Adenoviruses and Poxviruses, Rhabdoviruses, Paramixoviruses, Arenaviruses and Herpesviruses. We describe the rationale for, immunologic mechanisms involved in, and design of viral vectored gene vaccines under development and discuss the potential utility of these novel genetic vaccine approaches in eliciting protection against infectious diseases and cancer.
Collapse
Affiliation(s)
- Emanuele Sasso
- Nouscom srl, Via di Castel Romano 100, 00128 Rome, Italy; Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy.
| | | | - Nicola Zambrano
- Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Pansini 5, 80131 Naples, Italy.
| | | | | | - Alfredo Nicosia
- Ceinge-Biotecnologie Avanzate S.C. A.R.L., via Gaetano Salvatore 486, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University Federico II, Via Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
31
|
Diversify and Conquer: The Vaccine Escapism of Plasmodium falciparum. Microorganisms 2020; 8:microorganisms8111748. [PMID: 33171746 PMCID: PMC7694999 DOI: 10.3390/microorganisms8111748] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Over the last century, a great deal of effort and resources have been poured into the development of vaccines to protect against malaria, particularly targeting the most widely spread and deadly species of the human-infecting parasites: Plasmodium falciparum. Many of the known proteins the parasite uses to invade human cells have been tested as vaccine candidates. However, precisely because of the importance and immune visibility of these proteins, they tend to be very diverse, and in many cases redundant, which limits their efficacy in vaccine development. With the advent of genomics and constantly improving sequencing technologies, an increasingly clear picture is emerging of the vast genomic diversity of parasites from different geographic areas. This diversity is distributed throughout the genome and includes most of the vaccine candidates tested so far, playing an important role in the low efficacy achieved. Genomics is a powerful tool to search for genes that comply with the most desirable attributes of vaccine targets, allowing us to evaluate function, immunogenicity and also diversity in the worldwide parasite populations. Even predicting how this diversity might evolve and spread in the future becomes possible, and can inform novel vaccine efforts.
Collapse
|
32
|
Mo AXY, Pesce J, Augustine AD, Bodmer JL, Breen J, Leitner W, Hall BF. Understanding vaccine-elicited protective immunity against pre-erythrocytic stage malaria in endemic regions. Vaccine 2020; 38:7569-7577. [PMID: 33071001 DOI: 10.1016/j.vaccine.2020.09.071] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/26/2020] [Accepted: 09/24/2020] [Indexed: 12/17/2022]
Abstract
Recent malaria vaccine trials in endemic areas have yielded disparate results compared to studies conducted in non-endemic areas. A workshop was organized to discuss the differential pre-erythrocytic stage malaria vaccine (Pre-E-Vac) efficacies and underlying protective immunity under various conditions. It was concluded that many factors, including vaccine technology platforms, host genetics or physiologic conditions, and parasite and mosquito vector variations, may all contribute to Pre-E-Vac efficacy. Cross-disciplinary approaches are needed to decipher the multi-dimensional variables that contribute to the observed vaccine hypo-responsiveness. The malaria vaccine community has an opportunity to leverage recent advances in immunology, systems vaccinology, and high dimensionality data science methodologies to generate new clinical datasets with unprecedented levels of functional resolution as well as capitalize on existing datasets for comprehensive and aggregate analyses. These approaches would help to unlock our understanding of Pre-E-Vac immunology and to translate new candidates from the laboratory to the field more predictably.
Collapse
Affiliation(s)
- Annie X Y Mo
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA.
| | - John Pesce
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA
| | - Alison Deckhut Augustine
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA
| | | | - Joseph Breen
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA
| | - Wolfgang Leitner
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA
| | - B Fenton Hall
- National Institute of Allergy and Infectious Diseases, National Institute of Health, Department of Health and Human Service, Rockville, MD 20892, MSC 9825, USA
| |
Collapse
|
33
|
Abuga KM, Jones-Warner W, Hafalla JCR. Immune responses to malaria pre-erythrocytic stages: Implications for vaccine development. Parasite Immunol 2020; 43:e12795. [PMID: 32981095 PMCID: PMC7612353 DOI: 10.1111/pim.12795] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/26/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022]
Abstract
Radiation-attenuated sporozoites induce sterilizing immunity and remain the 'gold standard' for malaria vaccine development. Despite practical challenges in translating these whole sporozoite vaccines to large-scale intervention programmes, they have provided an excellent platform to dissect the immune responses to malaria pre-erythrocytic (PE) stages, comprising both sporozoites and exoerythrocytic forms. Investigations in rodent models have provided insights that led to the clinical translation of various vaccine candidates-including RTS,S/AS01, the most advanced candidate currently in a trial implementation programme in three African countries. With advances in immunology, transcriptomics and proteomics, and application of lessons from past failures, an effective, long-lasting and wide-scale malaria PE vaccine remains feasible. This review underscores the progress in PE vaccine development, focusing on our understanding of host-parasite immunological crosstalk in the tissue environments of the skin and the liver. We highlight possible gaps in the current knowledge of PE immunity that can impact future malaria vaccine development efforts.
Collapse
Affiliation(s)
- Kelvin Mokaya Abuga
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK.,Department of Epidemiology and Demography, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - William Jones-Warner
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Julius Clemence R Hafalla
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
34
|
Discovery of four new B-cell protective epitopes for malaria using Q beta virus-like particle as platform. NPJ Vaccines 2020; 5:92. [PMID: 33083027 PMCID: PMC7546618 DOI: 10.1038/s41541-020-00242-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/17/2020] [Indexed: 12/26/2022] Open
Abstract
Malaria remains one of the world’s most urgent global health problems, with almost half a million deaths and hundreds of millions of clinical cases each year. Existing interventions by themselves will not be enough to tackle infection in high-transmission areas. The best new intervention would be an effective vaccine; but the leading P. falciparum and P. vivax vaccine candidates, RTS,S and VMP001, show only modest to low field efficacy. New antigens and improved ways for screening antigens for protective efficacy will be required. This study exploits the potential of Virus-Like Particles (VLP) to enhance immune responses to antigens, the ease of coupling peptides to the Q beta (Qβ) VLP and the existing murine malaria challenge to screen B-cell epitopes for protective efficacy. We screened P. vivax TRAP (PvTRAP) immune sera against individual 20-mer PvTRAP peptides. The most immunogenic peptides associated with protection were loaded onto Qβ VLPs to assess protective efficacy in a malaria sporozoite challenge. A second approach focused on identifying conserved regions within known sporozoite invasion proteins and assessing them as part of the Qβ. Using this VLP as a peptide scaffold, four new protective B-cell epitopes were discovered: three from the disordered region of PvTRAP and one from Thrombospondin-related sporozoite protein (TRSP). Antigenic interference between these and other B-cell epitopes was also explored using the virus-like particle/peptide platform. This approach demonstrates the utility of VLPs to help identifying new B-cell epitopes for inclusion in next-generation malaria vaccines.
Collapse
|
35
|
Coughlan L. Factors Which Contribute to the Immunogenicity of Non-replicating Adenoviral Vectored Vaccines. Front Immunol 2020; 11:909. [PMID: 32508823 PMCID: PMC7248264 DOI: 10.3389/fimmu.2020.00909] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/20/2020] [Indexed: 01/12/2023] Open
Abstract
Adenoviral vectors are a safe and potently immunogenic vaccine delivery platform. Non-replicating Ad vectors possess several attributes which make them attractive vaccines for infectious disease, including their capacity for high titer growth, ease of manipulation, safety, and immunogenicity in clinical studies, as well as their compatibility with clinical manufacturing and thermo-stabilization procedures. In general, Ad vectors are immunogenic vaccines, which elicit robust transgene antigen-specific cellular (namely CD8+ T cells) and/or humoral immune responses. A large number of adenoviruses isolated from humans and non-human primates, which have low seroprevalence in humans, have been vectorized and tested as vaccines in animal models and humans. However, a distinct hierarchy of immunological potency has been identified between diverse Ad vectors, which unfortunately limits the potential use of many vectors which have otherwise desirable manufacturing characteristics. The precise mechanistic factors which underlie the profound disparities in immunogenicity are not clearly defined and are the subject of ongoing, detailed investigation. It has been suggested that a combination of factors contribute to the potent immunogenicity of particular Ad vectors, including the magnitude and duration of vaccine antigen expression following immunization. Furthermore, the excessive induction of Type I interferons by some Ad vectors has been suggested to impair transgene expression levels, dampening subsequent immune responses. Therefore, the induction of balanced, but not excessive stimulation of innate signaling is optimal. Entry factor binding or receptor usage of distinct Ad vectors can also affect their in vivo tropism following administration by different routes. The abundance and accessibility of innate immune cells and/or antigen-presenting cells at the site of injection contributes to early innate immune responses to Ad vaccination, affecting the outcome of the adaptive immune response. Although a significant amount of information exists regarding the tropism determinants of the common human adenovirus type-5 vector, very little is known about the receptor usage and tropism of rare species or non-human Ad vectors. Increased understanding of how different facets of the host response to Ad vectors contribute to their immunological potency will be essential for the development of optimized and customized Ad vaccine platforms for specific diseases.
Collapse
|
36
|
|
37
|
Modification of Adenovirus vaccine vector-induced immune responses by expression of a signalling molecule. Sci Rep 2020; 10:5716. [PMID: 32235848 PMCID: PMC7109070 DOI: 10.1038/s41598-020-61730-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/05/2020] [Indexed: 11/09/2022] Open
Abstract
Adenoviral vectors are being developed as vaccines against infectious agents and tumour-associated antigens, because of their ability to induce cellular immunity. However, the protection afforded in animal models has not easily translated into primates and clinical trials, underlying the need for improving adenoviral vaccines-induced immunogenicity. A Toll-like receptor signalling molecule, TRAM, was assessed for its ability to modify the immune responses induced by an adenovirus-based vaccine. Different adenovirus vectors either expressing TRAM alone or co-expressing TRAM along with a model antigen were constructed. The modification of T-cell and antibody responses induced by TRAM was assessed in vivo in mice and in primates. Co-expression of TRAM and an antigen from adenoviruses increased the transgene-specific CD8+ T cell responses in mice. Similar effects were seen when a TRAM expressing virus was co-administered with the antigen-expressing adenovirus. However, in primate studies, co-administration of a TRAM expressing adenovirus with a vaccine expressing the ME-TRAP malaria antigen had no significant effect on the immune responses. While these results support the idea that modification of innate immune signalling by genetic vectors modifies immunogenicity, they also emphasise the difficulty in generalising results from rodents into primates, where the regulatory pathway may be different to that in mice.
Collapse
|
38
|
Bettencourt P. Current Challenges in the Identification of Pre-Erythrocytic Malaria Vaccine Candidate Antigens. Front Immunol 2020; 11:190. [PMID: 32153565 PMCID: PMC7046804 DOI: 10.3389/fimmu.2020.00190] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/24/2020] [Indexed: 12/27/2022] Open
Abstract
Plasmodium spp.-infected mosquitos inject sporozoites into the skin of a mammalian host during a blood meal. These enter the host's circulatory system and establish an infection in the liver. After a silent metamorphosis, merozoites invade the blood leading to the symptomatic and transmissible stages of malaria. The silent pre-erythrocytic malaria stage represents a bottleneck in the disease which is ideal to block progression to clinical malaria, through chemotherapeutic and immunoprophylactic interventions. RTS,S/AS01, the only malaria vaccine close to licensure, although with poor efficacy, blocks the sporozoite invasion mainly through the action of antibodies against the CSP protein, a major component of the pellicle of the sporozoite. Strikingly, sterile protection against malaria can be obtained through immunization with radiation-attenuated sporozoites, genetically attenuated sporozoites or through chemoprophylaxis with infectious sporozoites in animals and humans, but the deployability of sporozoite-based live vaccines pose tremendous challenges. The protection induced by sporozoites occurs in the pre-erythrocytic stages and is mediated mainly by antibodies against the sporozoite and CD8+ T cells against peptides presented by MHC class I molecules in infected hepatocytes. Thus, the identification of malaria antigens expressed in the sporozoite and liver-stage may provide new vaccine candidates to be included, alone or in combination, as recombinant protein-based, virus-like particles or sub-unit virally-vectored vaccines. Here I review the efforts being made to identify Plasmodium falciparum antigens expressed during liver-stage with focus on the development of parasite, hepatocyte, mouse models, and resulting rate of infection in order to identify new vaccine candidates and to improve the efficacy of the current vaccines. Finally, I propose new approaches for the identification of liver-stage antigens based on immunopeptidomics.
Collapse
|
39
|
Khaemba EN, Ogwang C, Kinyanjui S, Muindi JM, Koske JK, Kimani D, Ngoi J, Mwacharo J, Shangala J, Njuguna P, Mutinda D, Nyatichi E, Peshu J, Mutinda B, Ndungu FM, Farnert A, Bashraheil MM, Bejon P, Kapulu MC. Comparing drug regimens for clearance of malaria parasites in asymptomatic adults using PCR in Kilifi County, Kenya: an open-label randomised controlled clinical trial (MalPaC). Wellcome Open Res 2020. [DOI: 10.12688/wellcomeopenres.15627.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: To restrict trial endpoints to infections acquired after vaccination in Phase IIb trials of candidate malaria vaccines, participants are treated with anti-malarial drugs to clear existing infections. Anti-malarial drugs with a long half-life may inhibit the acquisition of new infections. This study evaluated the effects of three anti-malarial drug regimens on the clearance of existing infections and acquisition of new infections. Methods: An open-label randomised controlled trial (MalPaC) was conducted between November 2013 and February 2014. Ninety adults were randomised 1:1:1 to receive one of three treatments: atovaquone/proguanil and artesunate (AP+AS); artesunate (AS); or sulphadoxine-pyrimethamine, artesunate, and primaquine (SP+AS+PQ). Parasite monitoring was determined over 84-day follow-up by assessing Plasmodium falciparum positivity by 18s qPCR, live and sexual stage parasites by RT-PCR, and recrudescence of infections by msp2 genotyping. Results: At enrolment, parasite prevalence by qPCR was 44% (40/90, day 0), which fell to 10% (9/90, day 16), then rose to almost the initial rates by day 84 (39%, 35/90). Individuals treated with AS and SP+AS+PQ were more likely to have higher qPCR positive rates compared to participants treated with AP+AS in the immediate post-treatment phase (days 16-28) (OR=7.7 [95%CI 4.6-12.8] p<0.0005 and OR=4.2 [95%CI 2.6-6.8] p<0.0005, respectively). In the immediate post-treatment phase, qPCR positivity was less likely associated with evidence of live parasites and gametocytaemia. Prevalence of “old”, “new” or “undetectable” infections did not differ significantly over time or drug regimen. However, participants on the AP+AS drug regimen were less likely to have parasite infection recrudescence compared to participants treated with AS and SP+AS+PQ. Conclusion: Falciparum DNA remained detectable by PCR post-treatment with incomplete parasite clearance regardless of drug regimen. Though AP+AS drug regimen may also have partially suppressed the acquisition of new infections during post-treatment follow-up. Trial registration: Pan African Clinical Trials Registry, 22nd of August 2013, PACTR201309000625311.
Collapse
|
40
|
Molina-Franky J, Cuy-Chaparro L, Camargo A, Reyes C, Gómez M, Salamanca DR, Patarroyo MA, Patarroyo ME. Plasmodium falciparum pre-erythrocytic stage vaccine development. Malar J 2020; 19:56. [PMID: 32013956 PMCID: PMC6998842 DOI: 10.1186/s12936-020-3141-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 01/25/2020] [Indexed: 12/13/2022] Open
Abstract
Worldwide strategies between 2010 and 2017 aimed at controlling malarial parasites (mainly Plasmodium falciparum) led to a reduction of just 18% regarding disease incidence rates. Many biologically-derived anti-malarial vaccine candidates have been developed to date; this has involved using many experimental animals, an immense amount of work and the investment of millions of dollars. This review provides an overview of the current state and the main results of clinical trials for sporozoite-targeting vaccines (i.e. the parasite stage infecting the liver) carried out by research groups in areas having variable malaria transmission rates. However, none has led to promising results regarding the effective control of the disease, thereby making it necessary to complement such efforts at finding/introducing new vaccine candidates by adopting a multi-epitope, multi-stage approach, based on minimal subunits of the main sporozoite proteins involved in the invasion of the liver.
Collapse
Affiliation(s)
- Jessica Molina-Franky
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Laura Cuy-Chaparro
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Anny Camargo
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - César Reyes
- PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia.,Animal Science Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Bogotá, Colombia.,3D Structures Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Marcela Gómez
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - David Ricardo Salamanca
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia.,Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Program in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia. .,Basic Sciences Department, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.
| | - Manuel Elkin Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia. .,Medical School, Universidad Nacional de Colombia, Bogotá, Colombia.
| |
Collapse
|
41
|
Preclinical Development and Assessment of Viral Vectors Expressing a Fusion Antigen of Plasmodium falciparum LSA1 and LSAP2 for Efficacy against Liver-Stage Malaria. Infect Immun 2020; 88:IAI.00573-19. [PMID: 31740525 PMCID: PMC6977128 DOI: 10.1128/iai.00573-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
Despite promising progress in malaria vaccine development in recent years, an efficacious subunit vaccine against Plasmodium falciparum remains to be licensed and deployed. Cell-mediated protection from liver-stage malaria relies on a sufficient number of antigen-specific T cells reaching the liver during the time that parasites are present. A single vaccine expressing two antigens could potentially increase both the size and breadth of the antigen-specific response while halving vaccine production costs. Despite promising progress in malaria vaccine development in recent years, an efficacious subunit vaccine against Plasmodium falciparum remains to be licensed and deployed. Cell-mediated protection from liver-stage malaria relies on a sufficient number of antigen-specific T cells reaching the liver during the time that parasites are present. A single vaccine expressing two antigens could potentially increase both the size and breadth of the antigen-specific response while halving vaccine production costs. In this study, we investigated combining two liver-stage antigens, P. falciparum LSA1 (PfLSA1) and PfLSAP2, and investigated the induction of protective efficacy by coadministration of single-antigen vectors or vaccination with dual-antigen vectors, using simian adenovirus and modified vaccinia virus Ankara vectors. The efficacy of these vaccines was assessed in mouse malaria challenge models using chimeric P. berghei parasites expressing the relevant P. falciparum antigens and challenging mice at the peak of the T cell response. Vaccination with a combination of the single-antigen vectors expressing PfLSA1 or PfLSAP2 was shown to improve protective efficacy compared to vaccination with each single-antigen vector alone. Vaccination with dual-antigen vectors expressing both PfLSA1 and PfLSAP2 resulted in responses to both antigens, particularly in outbred mice, and most importantly, the efficacy was equivalent to that of vaccination with a mixture of single-antigen vectors. Based on these promising data, dual-antigen vectors expressing PfLSA1 and PfLSAP2 will now proceed to manufacturing and clinical assessment under good manufacturing practice (GMP) guidelines.
Collapse
|
42
|
Design and assessment of TRAP-CSP fusion antigens as effective malaria vaccines. PLoS One 2020; 15:e0216260. [PMID: 31967991 PMCID: PMC6975556 DOI: 10.1371/journal.pone.0216260] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 12/16/2019] [Indexed: 11/25/2022] Open
Abstract
The circumsporozoite protein (CSP) and thrombospondin-related adhesion protein (TRAP) are major targets for pre-erythrocytic malaria vaccine development. However, the CSP-based vaccine RTS,S provides only marginal protection, highlighting the need for innovative vaccine design and development. Here we design and characterize expression and folding of P. berghei (Pb) and P. falciparum (Pf) TRAP-CSP fusion proteins, and evaluate immunogenicity and sterilizing immunity in mice. TRAP N-terminal domains were fused to the CSP C-terminal αTSR domain with or without the CSP repeat region, expressed in mammalian cells, and evaluated with or without N-glycan shaving. Pb and Pf fusions were each expressed substantially better than the TRAP or CSP components alone; furthermore, the fusions but not the CSP component could be purified to homogeneity and were well folded and monomeric. As yields of TRAP and CSP fragments were insufficient, we immunized BALB/c mice with Pb TRAP-CSP fusions in AddaVax adjuvant and tested the effects of absence or presence of the CSP repeats and absence or presence of high mannose N-glycans on total antibody titer and protection from infection by mosquito bite both 2.5 months and 6 months after the last immunization. Fusions containing the repeats were completely protective against challenge and re-challenge, while those lacking repeats were significantly less effective. These results correlated with higher total antibody titers when repeats were present. Our results show that TRAP-CSP fusions increase protein antigen production, have the potential to yield effective vaccines, and also guide design of effective proteins that can be encoded by nucleic acid-based and virally vectored vaccines.
Collapse
|
43
|
Wilkie M, Satti I, Minhinnick A, Harris S, Riste M, Ramon RL, Sheehan S, Thomas ZRM, Wright D, Stockdale L, Hamidi A, O'Shea MK, Dwivedi K, Behrens HM, Davenne T, Morton J, Vermaak S, Lawrie A, Moss P, McShane H. A phase I trial evaluating the safety and immunogenicity of a candidate tuberculosis vaccination regimen, ChAdOx1 85A prime - MVA85A boost in healthy UK adults. Vaccine 2019; 38:779-789. [PMID: 31735500 PMCID: PMC6985898 DOI: 10.1016/j.vaccine.2019.10.102] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 11/18/2022]
Abstract
Background This phase I trial evaluated the safety and immunogenicity of a candidate tuberculosis vaccination regimen, ChAdOx1 85A prime-MVA85A boost, previously demonstrated to be protective in animal studies, in healthy UK adults. Methods We enrolled 42 healthy, BCG-vaccinated adults into 4 groups: low dose Starter Group (n = 6; ChAdOx1 85A alone), high dose groups; Group A (n = 12; ChAdOx1 85A), Group B (n = 12; ChAdOx1 85A prime – MVA85A boost) or Group C (n = 12; ChAdOx1 85A – ChAdOx1 85A prime – MVA85A boost). Safety was determined by collection of solicited and unsolicited vaccine-related adverse events (AEs). Immunogenicity was measured by antigen-specific ex-vivo IFN-γ ELISpot, IgG serum ELISA, and antigen-specific intracellular IFN-γ, TNF-α, IL-2 and IL-17. Results AEs were mostly mild/moderate, with no Serious Adverse Events. ChAdOx1 85A induced Ag85A-specific ELISpot and intracellular cytokine CD4+ and CD8+ T cell responses, which were not boosted by a second dose, but were boosted with MVA85A. Polyfunctional CD4+ T cells (IFN-γ, TNF-α and IL-2) and IFN-γ+, TNF-α+ CD8+ T cells were induced by ChAdOx1 85A and boosted by MVA85A. ChAdOx1 85A induced serum Ag85A IgG responses which were boosted by MVA85A. Conclusion A ChAdOx1 85A prime – MVA85A boost is well tolerated and immunogenic in healthy UK adults.
Collapse
Affiliation(s)
- Morven Wilkie
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Iman Satti
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | | | - Michael Riste
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Sharon Sheehan
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Daniel Wright
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Lisa Stockdale
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Ali Hamidi
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Kritica Dwivedi
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Tamara Davenne
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Joshua Morton
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Alison Lawrie
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| |
Collapse
|
44
|
Cable J, Srikantiah P, Crowe JE, Pulendran B, Hill A, Ginsberg A, Koff W, Mathew A, Ng T, Jansen K, Glenn G, Permar S, Wilson I, Weiner DB, Weissman D, Rappuoli R. Vaccine innovations for emerging infectious diseases-a symposium report. Ann N Y Acad Sci 2019; 1462:14-26. [PMID: 31659752 DOI: 10.1111/nyas.14235] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 08/28/2019] [Indexed: 12/29/2022]
Abstract
Vaccines have been incredibly successful at stemming the morbidity and mortality of infectious diseases worldwide. However, there are still no effective vaccines for many serious and potentially preventable infectious diseases. Advances in vaccine technology, including new delivery methods and adjuvants, as well as progress in systems biology and an increased understanding of the human immune system, hold the potential to address these issues. In addition, maternal immunization has opened an avenue to address infectious diseases in neonates and very young infants. This report summarizes the presentations from a 1-day symposium at the New York Academy of Sciences entitled "Innovative Vaccines against Resistant Infectious Diseases and Emerging Threats," held on May 20, 2019.
Collapse
Affiliation(s)
| | | | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center; and Vanderbilt University, Nashville, Tennessee
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection; Department of Pathology; and Department of Microbiology & Immunology, Stanford University, Stanford, California
| | - Adrian Hill
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Ann Ginsberg
- International AIDS Vaccine Initiative, New York, New York
| | - Wayne Koff
- The Human Vaccines Project, New York, New York
| | - Anuja Mathew
- Department of Cell and Molecular Biology, The University of Rhode Island, Kingston, Rhode Island
| | - Tony Ng
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, New York, New York
| | | | | | | | - Ian Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, San Diego, California
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania
| | - Drew Weissman
- Department of Medicine, The University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | | |
Collapse
|
45
|
Matos ADS, Rodrigues-da-Silva RN, Soares IF, Baptista BDO, de Souza RM, Bitencourt-Chaves L, Totino PRR, Sánchez-Arcila JC, Daniel-Ribeiro CT, López-Camacho C, Reyes-Sandoval A, Pratt-Riccio LR, Lima-Junior JDC. Antibody Responses Against Plasmodium vivax TRAP Recombinant and Synthetic Antigens in Naturally Exposed Individuals From the Brazilian Amazon. Front Immunol 2019; 10:2230. [PMID: 31620136 PMCID: PMC6763564 DOI: 10.3389/fimmu.2019.02230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/03/2019] [Indexed: 11/13/2022] Open
Abstract
Thrombospondin-related adhesive protein (TRAP) is essential for sporozoite motility and the invasion of mosquitoes' salivary gland and vertebrate's hepatocyte and is, thus, considered a promising pre-erythrocytic vaccine candidate. Despite the existence of a few reports on naturally acquired immune response against Plasmodium vivax TRAP (PvTRAP), it has never been explored so far in the Amazon region, so results are conflicting. Here, we characterized the (IgG and IgG subclass) antibody reactivity against recombinant PvTRAP in a cross-sectional study of 299 individuals exposed to malaria infection in three municipalities (Cruzeiro do Sul, Mâncio Lima and Guajará) from the Acre state of the Brazilian Amazon. In addition, the full PvTRAP sequence was screened for B-cell epitopes using in silico and in vitro approaches. Firstly, we confirmed that PvTRAP is naturally immunogenic in the cohort population since 49% of the individuals were IgG-responders to it. The observed immune responses were mainly driven by cytophilic IgG1 over all other sublcasses and the IgG levels that was corelated with age and time of residence in the studied area (p < 0.05). Interestingly, only the levels of specific anti-TRAP IgG3 seemed to be associated with protection, as IgG3 responders presented a significantly higher time elapse since the last malaria episode than those recorded for IgG3 non-responders. Regarding the B-cell epitope mapping, among the 148 responders to PvTRAP, four predicted epitopes were confirmed by recognition of antibodies (PvTRAPR197-H227; PvTRAPE237-T258; PvTRAPP344-G374; and PvTRAPE439-K454). Nevertheless, the frequency of responders against these peptides were low and did not show a clear correlation with the antibody response against the corresponding antigen. Moreover, none of the linear confirmed epitopes were located in the binding regions of PvTRAP in respect to the host cell ligand. Collectively, our data confirm the PvTRAP immunogenicity among Amazon inhabitants, while suggesting that the main important B-cell epitopes are not linear.
Collapse
Affiliation(s)
- Ada da Silva Matos
- Laboratory of Immunoparasitology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | | | | | | | | | - Lana Bitencourt-Chaves
- Laboratory of Immunoparasitology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | | | - Juan Camilo Sánchez-Arcila
- Viral Immunology Laboratory, Oswaldo Cruz Institute, IOC, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | | | - César López-Camacho
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Arturo Reyes-Sandoval
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | |
Collapse
|
46
|
Dejon-Agobe JC, Ateba-Ngoa U, Lalremruata A, Homoet A, Engelhorn J, Nouatin OP, Edoa JR, Fernandes JF, Esen M, Mouwenda YD, Betouke Ongwe EM, Massinga-Loembe M, Hoffman SL, Sim BKL, Theisen M, Kremsner PG, Adegnika AA, Lell B, Mordmüller B. Controlled Human Malaria Infection of Healthy Adults With Lifelong Malaria Exposure to Assess Safety, Immunogenicity, and Efficacy of the Asexual Blood Stage Malaria Vaccine Candidate GMZ2. Clin Infect Dis 2019; 69:1377-1384. [PMID: 30561539 PMCID: PMC6763635 DOI: 10.1093/cid/ciy1087] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/18/2018] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND GMZ2 is a recombinant malaria vaccine inducing immune responses against Plasmodium falciparum (Pf) merozoite surface protein-3 and glutamate-rich protein. We used standardized controlled human malaria infection (CHMI) to assess the efficacy of this asexual blood-stage vaccine. METHODS We vaccinated 50 healthy, adult volunteers with lifelong exposure to Pf 3 times, at 4-week intervals, with 30 or 100 µg GMZ2 formulated in CAF01, a liposome-based adjuvant; 100 µg GMZ2, formulated in Alhydrogel; or a control vaccine (Verorab). Approximately 13 weeks after the last vaccination, 35/50 volunteers underwent CHMI by direct venous inoculation of 3200 Pf sporozoites (Sanaria® PfSPZ Challenge). RESULTS Adverse events were similarly distributed between GMZ2 and control vaccinees. Baseline-corrected anti-GMZ2 antibody concentrations 4 weeks after the last vaccination were higher in all 3 GMZ2-vaccinated arms, compared to the control group. All GMZ2 formulations induced similar antibody levels. CHMI resulted in 29/34 (85%) volunteers with Pf parasitemia and 15/34 (44%) with malaria (parasitemia and symptoms). The proportion of participants with malaria (2/5 control, 6/10 GMZ2-Alhydrogel, 2/8 30 µg GMZ2-CAF01, and 5/11 100 µg GMZ2-CAF01) and the time it took them to develop malaria were similar in all groups. Baseline, vaccine-specific antibody concentrations were associated with protection against malaria. CONCLUSIONS GMZ2 is well tolerated and immunogenic in lifelong-Pf-exposed adults from Gabon, with similar antibody responses regardless of formulation. CHMI showed no protective effect of prior vaccination with GMZ2, although baseline, vaccine-specific antibody concentrations were associated with protection. CHMI with the PfSPZ Challenge is a potent new tool to validate asexual, blood-stage malaria vaccines in Africa. CLINICAL TRIALS REGISTRATION Pan-African Clinical Trials: PACTR201503001038304.
Collapse
Affiliation(s)
- Jean Claude Dejon-Agobe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Ulysse Ateba-Ngoa
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Albert Lalremruata
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Andreas Homoet
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Julie Engelhorn
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Odilon Paterne Nouatin
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Jean Ronald Edoa
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - José F Fernandes
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Meral Esen
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Yoanne Darelle Mouwenda
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Eunice M Betouke Ongwe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
| | - Marguerite Massinga-Loembe
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | | | | | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Denmark
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Ayôla A Adegnika
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
- Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné and African Partner Institution, German Center for Infection Research, Gabon
- Institut für Tropenmedizin, Universität Tübingen and German Center for Infection Research, Germany
| |
Collapse
|
47
|
Abstract
Malaria vaccine development has rapidly advanced in the past decade. The very first phase 3 clinical trial of the RTS,S vaccine was completed with over 15,000 African infants and children, and pilot implementation studies are underway. Next-generation candidate vaccines using novel antigens, platforms, or approaches targeting different and/or multiple stages of the Plasmodium life cycle are being tested. Many candidates, in various stages of development, promise enhanced efficacy of long duration and broad protection against genetically diverse malaria strains, with a few studies under way in target populations in endemic areas. Malaria vaccines together with other interventions promise interruption and eventual elimination of malaria in endemic areas.
Collapse
Affiliation(s)
- Matthew B Laurens
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore 21201, Maryland, USA; .,Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.,Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore 21201, Maryland, USA
| |
Collapse
|
48
|
Martin MD, Jensen IJ, Ishizuka AS, Lefebvre M, Shan Q, Xue HH, Harty JT, Seder RA, Badovinac VP. Bystander responses impact accurate detection of murine and human antigen-specific CD8 T cells. J Clin Invest 2019; 129:3894-3908. [PMID: 31219804 DOI: 10.1172/jci124443] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Induction of memory CD8 T cells is important for controlling infections such as malaria HIV/AIDS, and for cancer immunotherapy. Accurate assessment of antigen (Ag)-specific CD8 T-cells is critical for vaccine optimization and defining correlates of protection. However, conditions for determining Ag-specific CD8 T-cell responses ex-vivo using ICS may be variable, especially in humans with complex antigens. Here, we used an attenuated whole parasite malaria vaccine model in humans and various experimental infections in mice to show that the duration of antigenic stimulation and timing of brefeldin A (BFA) addition influences the magnitude of Ag-specific and bystander T cell responses. Indeed, following immunization with an attenuated whole sporozoite malaria vaccine in humans, significantly higher numbers of IFN-γ producing memory CD8 T-cells comprised of antigen specific and bystander responses were detected by increasing the duration of Ag-stimulation prior to addition of BFA. Mechanistic analyses of virus-specific CD8 T-cells in mice revealed that the increase in IFNg producing CD8 T-cells was due to bystander activation of Ag-experienced memory CD8 T-cells, and correlated with the proportion of Ag-experienced CD8 T-cells in the stimulated populations. Incubation with anti-cytokine antibodies (ex. IL-12) improved accuracy in detecting bona-fide memory CD8 T-cell responses suggesting this as the mechanism for the bystander activation. These data have important implications for accurate assessment of immune responses generated by vaccines intended to elicit protective memory CD8 T-cells.
Collapse
Affiliation(s)
| | - Isaac J Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Andrew S Ishizuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Mitchell Lefebvre
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Qiang Shan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Hai-Hui Xue
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA.,Iowa City Veterans Affairs Health Care System, Iowa City, Iowa, USA
| | - John T Harty
- Department of Pathology and.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Vladimir P Badovinac
- Department of Pathology and.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
49
|
Goh YS, McGuire D, Rénia L. Vaccination With Sporozoites: Models and Correlates of Protection. Front Immunol 2019; 10:1227. [PMID: 31231377 PMCID: PMC6560154 DOI: 10.3389/fimmu.2019.01227] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Despite continuous efforts, the century-old goal of eradicating malaria still remains. Multiple control interventions need to be in place simultaneously to achieve this goal. In addition to effective control measures, drug therapies and insecticides, vaccines are critical to reduce mortality and morbidity. Hence, there are numerous studies investigating various malaria vaccine candidates. Most of the malaria vaccine candidates are subunit vaccines. However, they have shown limited efficacy in Phase II and III studies. To date, only whole parasite formulations have been shown to induce sterile immunity in human. In this article, we review and discuss the recent developments in vaccination with sporozoites and the mechanisms of protection involved.
Collapse
Affiliation(s)
- Yun Shan Goh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore
| | - Daniel McGuire
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
50
|
Mehrizi AA, Ameri Torzani M, Zakeri S, Jafary Zadeh A, Babaeekhou L. Th1 immune response to Plasmodium falciparum recombinant thrombospondin-related adhesive protein (TRAP) antigen is enhanced by TLR3-specific adjuvant, poly(I:C) in BALB/c mice. Parasite Immunol 2019; 40:e12538. [PMID: 29799636 DOI: 10.1111/pim.12538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 05/21/2018] [Indexed: 12/12/2022]
Abstract
Sporozoite-based malaria vaccines have provided a gold standard for malaria vaccine development, and thrombospondin-related adhesive protein (TRAP) serves as the main vaccine candidate antigen on sporozoites. As recombinant malaria vaccine candidate antigens are poorly immunogenic, additional appropriate immunostimulants, such as an efficient adjuvant, are highly essential to modulate Th1-cell predominance and also to induce a protective and long-lived immune response. In this study, polyinosinic:polycytidylic acid [poly(I:C)], the ligand of TLR3, was considered as the potential adjuvant for vaccines targeting stronger Th1-based immune responses. For this purpose, BALB/c mice were immunized with rPfTRAP delivered in putative poly(I:C) adjuvant, and humoural and cellular immune responses were determined in different immunized mouse groups. Delivery of rPfTRAP with poly(I:C) induced high levels and titres of persisted and also high-avidity anti-rPfTRAP IgG antibodies comparable to complete Freund's adjuvant (CFA)/incomplete Freund's adjuvant (IFA) adjuvant after the second boost. In addition, rPfTRAP formulated with poly(I:C) elicited a higher ratio of IFN-γ/IL-5, IgG2a/IgG1, and IgG2b/IgG1 than with CFA/IFA, indicating that poly(I:C) supports the induction of a stronger Th1-based immune response. This is a first time study which reveals the potential of rPfTRAP delivery in poly(I:C) to increase the level, avidity and durability of both anti-PfTRAP cytophilic antibodies and Th1 cytokines.
Collapse
Affiliation(s)
- A A Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - M Ameri Torzani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.,Department of Biology, Islamshahr Branch, Islamic Azad University, Islamshahr, Iran
| | - S Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - A Jafary Zadeh
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - L Babaeekhou
- Department of Biology, Islamshahr Branch, Islamic Azad University, Islamshahr, Iran
| |
Collapse
|