1
|
Levy O. Vaccinology: Getting our modernization act together. J Exp Med 2025; 222:e20240961. [PMID: 40261250 PMCID: PMC12013515 DOI: 10.1084/jem.20240961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025] Open
Abstract
Ofer Levy, Director, Precision Vaccines Program at Boston Children's Hospital, reflects on implications of the new FDA Modernization Act 2.0 on accelerating drug and vaccine discovery and development.
Collapse
Affiliation(s)
- Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
2
|
Nassuuna J, Sterk J, Walusimbi B, Natukunda A, Nkangi R, Amongin R, Zirimenya L, Webb EL, Elliott AM, Nkurunungi G. Helminth driven gut inflammation and microbial translocation associate with altered vaccine responses in rural Uganda. NPJ Vaccines 2025; 10:56. [PMID: 40140378 PMCID: PMC11947158 DOI: 10.1038/s41541-025-01116-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Vaccine responses are sometimes impaired in rural, low-income settings. Helminth-associated gut barrier dysfunction and microbial translocation (MT) may be implicated. We used samples from a trial of praziquantel treatment-effects on vaccine responses in Schistosoma mansoni (Sm)-endemic Ugandan islands, measuring intestinal fatty acid-binding protein 2 (I-FABP2), lipopolysaccharide-binding protein, anti-endotoxin core antibodies (EndoCab), soluble CD14 (sCD14) in plasma, and faecal lipocalin-2, occult blood (FOB), and calprotectin (fCAL), and evaluating their associations with baseline helminth infection, praziquantel treatment, and responses to BCG, yellow fever, typhoid, HPV, and tetanus-diphtheria vaccines. Sm associated positively with fCAL and FOB, hookworm with I-FABP2, and any helminth with EndoCab IgM, fCAL and FOB. Sm associated inversely with sCD14. Praziquantel treatment reduced all marker concentrations, significantly fCAL and FOB, implying that Sm-associated gut inflammation and MT is reversible. Associations of assessed markers with vaccine-specific responses were predominantly inverse. Interventions to improve gut barrier function may enhance vaccine responsiveness.
Collapse
Affiliation(s)
- Jacent Nassuuna
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | | | - Bridgious Walusimbi
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Agnes Natukunda
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- International Statistics and Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Ronald Nkangi
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, The Netherlands
| | - Rebecca Amongin
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Ludoviko Zirimenya
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Emily L Webb
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- International Statistics and Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Alison M Elliott
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Gyaviira Nkurunungi
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda.
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
3
|
Gong XY, Wei JCC, Zhang LY. Possible perspectives for cardiovascular events following coronavirus disease 2019 vaccination. Eur Heart J 2025:ehaf112. [PMID: 40053691 DOI: 10.1093/eurheartj/ehaf112] [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: 03/09/2025] Open
Affiliation(s)
- Xue-Yan Gong
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, No.99 Longcheng Street, Taiyuan, Shanxi 030032, China
| | - James Cheng-Chung Wei
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, No.99 Longcheng Street, Taiyuan, Shanxi 030032, China
- Institute of Medicine, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., South District, Taichung 40201, Taiwan
- Department of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Rd., South District, Taichung 40201, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, No. 110, Sec. 1, Jianguo N. Rd., South District, Taichung 40201, Taiwan
| | - Li-Yun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, No.99 Longcheng Street, Taiyuan, Shanxi 030032, China
| |
Collapse
|
4
|
Mbow M, Hoving D, Cisse M, Diallo I, Honkpehedji YJ, Huisman W, Pothast CR, Jongsma MLM, König MH, de Kroon AC, Linh LTK, Azimi S, Tak T, Kruize YCM, Kurniawan F, Dia YA, Zhang JLH, Prins C, Roukens AHE, de Vries JJC, Wammes LJ, Smits HH, Adegnika AA, Zlei M, Kuijpers TW, Wieske L, Dieye A, Mboup S, Kremsner PG, Eftimov F, Velavan TP, Berlin I, Heemskerk MHM, Yazdanbakhsh M, Jochems SP. Immune responses to SARS-CoV-2 in sub-Saharan Africa and western Europe: a retrospective, population-based, cross-sectional study. THE LANCET. MICROBE 2025; 6:100942. [PMID: 39708825 DOI: 10.1016/j.lanmic.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/17/2024] [Accepted: 07/03/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND SARS-CoV-2 has been associated with a higher proportion of asymptomatic infections and lower mortality in sub-Saharan Africa than high-income countries. However, there is currently a lack of data on cellular immune responses to SARS-CoV-2 in people living in Africa compared with people in high-income regions of the world. We aimed to assess geographical variation in peripheral and mucosal immune responses. METHODS In this retrospective, population-based, cross-sectional study, we analysed peripheral blood and nasal curettage samples from seven clinical studies involving individuals from Senegal (Senegalese cohort), the Netherlands, and Germany (European cohort). Samples were collected between Nov 1, 2018, and Dec 20, 2021. We included samples from individuals with no, mild, or severe COVID-19. A validation cohort of individuals from Senegal and Gabon (n=64) was used to validate key findings from the main cohort. Matching of individuals between geographical regions by age, sex, viral load, and infection severity and duration was used to address confounding factors. We examined the cellular, humoral, and cytokine immune responses using cytometry by time of flight, spectral flow cytometry, ELISA, and Luminex. FINDINGS We included 133 individuals (59 from the Senegalese cohort and 74 from the European cohort). In contrast to the European cohort, mild COVID-19 in the Senegalese cohort was not associated with any statistically significant perturbations in blood or nasal immune cell profiles, nor with increased pro-inflammatory cytokines, although SARS-CoV-2-specific adaptive immunity was readily induced, as seen in Europeans. In severe COVID-19, both the Senegalese and European cohorts showed lymphopenia (Senegal: 2·9-times decrease, p=0·0010 vs Europe: 1·6-times decrease, p=0·0046) and increased neutrophil frequencies in blood (Senegal: 2·0-times increase, p=0·0044 vs Europe: 1·3-times increase, p=0·026) and the nasal mucosa CD66b+CD16low neutrophils (Senegal: 9·9-times increase, p=0·045 vs Europe: 392-times increase, p<0·0001). However, in contrast to Europeans, the Senegalese cohort had no significant expansion of immature immune populations, inflammasome activation, or monocyte recruitment to the nasal mucosa. INTERPRETATION The observed divergent immunological trajectories during SARS-CoV-2 infection offer a potential explanation for the reported attenuated disease course in sub-Saharan Africa and highlight the need to further investigate immune responses to SARS-CoV-2 in understudied populations. FUNDING European and Developing Countries Clinical Trials Partnership 2 programme (AIDCO), LUMC Gisela Thier Fellowship, Dutch Research Council (NWO), European Research Council, and Leids Universitair Fonds.
Collapse
Affiliation(s)
- Moustapha Mbow
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal; Institute of Health Research, Epidemiological Surveillance and Training, Dakar, Senegal
| | - Dennis Hoving
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Marouba Cisse
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal; Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Ibrahima Diallo
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal; Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Yabo J Honkpehedji
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Wesley Huisman
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Cilia R Pothast
- Department of Hematology Leiden University Medical Centre, Leiden, Netherlands
| | - Marlieke L M Jongsma
- Department of Cell and Chemical Biology, ONCODE Institute Leiden University Medical Centre, Leiden, Netherlands
| | - Marion H König
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Alicia C de Kroon
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Le Thi Kieu Linh
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany; Vietnamese-German Center for Medical Research, VG-CARE, Hanoi, Viet Nam
| | - Shohreh Azimi
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Tamar Tak
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Yvonne C M Kruize
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Farid Kurniawan
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands; Division of Endocrinology, Metabolism, and Diabetes, Department of Internal Medicine, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Yacine Amet Dia
- Institute of Health Research, Epidemiological Surveillance and Training, Dakar, Senegal
| | - Jaimie L H Zhang
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Corine Prins
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Anna H E Roukens
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Jutte J C de Vries
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Linda J Wammes
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Hermelijn H Smits
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Ayola A Adegnika
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon; Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany; The German Center for Infection Research (DZIF), Tübingen, Germany
| | - Mihaela Zlei
- Department of Immunology Leiden University Medical Centre, Leiden, Netherlands; Department of Flow Cytometry, Regional Institute of Oncology, Iasi, Romania
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Disease Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Luuk Wieske
- Department of Neurology and Neurophysiology Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Alioune Dieye
- Department of Immunology, Faculty of Medicine, Pharmacy, and Odontology, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| | - Souleymane Mboup
- Institute of Health Research, Epidemiological Surveillance and Training, Dakar, Senegal
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon; Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany; The German Center for Infection Research (DZIF), Tübingen, Germany
| | - Filip Eftimov
- Department of Neurology and Neurophysiology Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Thirumalaisamy P Velavan
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany; The German Center for Infection Research (DZIF), Tübingen, Germany; Vietnamese-German Center for Medical Research, VG-CARE, Hanoi, Viet Nam; Faculty of Medicine, Duy Tan University, Da Nang, Viet Nam
| | - Ilana Berlin
- Department of Cell and Chemical Biology, ONCODE Institute Leiden University Medical Centre, Leiden, Netherlands
| | | | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands
| | - Simon P Jochems
- Leiden University Center for Infectious Diseases Leiden University Medical Centre, Leiden, Netherlands.
| |
Collapse
|
5
|
Ruzzi F, Riccardo F, Conti L, Tarone L, Semprini MS, Bolli E, Barutello G, Quaglino E, Lollini PL, Cavallo F. Cancer vaccines: Target antigens, vaccine platforms and preclinical models. Mol Aspects Med 2025; 101:101324. [PMID: 39631227 DOI: 10.1016/j.mam.2024.101324] [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: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
This review provides a comprehensive overview of the evolving landscape of cancer vaccines, highlighting their potential to revolutionize tumor prevention. Building on the success of vaccines against virus-related cancers, such as HPV- and HBV-associated cervical and liver cancers, the current challenge is to extend these achievements to the prevention of non-viral tumors and the treatment of preneoplastic or early neoplastic lesions. This review analyzes the critical aspects of preventive anti-cancer vaccination, focusing on the choice of target antigens, the development of effective vaccine platforms and technologies, and the use of various model systems for preclinical testing, from laboratory rodents to companion animals.
Collapse
Affiliation(s)
- Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Federica Riccardo
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Laura Conti
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Lidia Tarone
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Maria Sofia Semprini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Elisabetta Bolli
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Giuseppina Barutello
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Elena Quaglino
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy; IRCCS Azienda Ospedaliera Universitaria di Bologna, 40138, Bologna, Italy.
| | - Federica Cavallo
- Laboratory of OncoImmunology, Department of Molecular Biotechnology and Health Sciences (DMBSS), University of Torino, 10126, Torino, Italy.
| |
Collapse
|
6
|
Chayé MAM, van Hengel ORJ, Voskamp AL, Ozir-Fazalalikhan A, König MH, Stam KA, Manurung MD, Mouwenda YD, Aryeetey YA, Kurniawan A, Kruize YCM, Sartono E, Buisman AM, Yazdanbakhsh M, Tak T, Smits HH. Multi-dimensional analysis of B cells reveals the expansion of memory and regulatory B-cell clusters in humans living in rural tropical areas. Clin Exp Immunol 2025; 219:uxae074. [PMID: 39129562 PMCID: PMC11771192 DOI: 10.1093/cei/uxae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 07/06/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024] Open
Abstract
B-cells play a critical role in the formation of immune responses against pathogens by acting as antigen-presenting cells, by modulating immune responses, and by generating immune memory and antibody responses. Here, we studied B-cell subset distributions between regions with higher and lower microbial exposure, i.e. by comparing peripheral blood B-cells from people living in Indonesia or Ghana to those from healthy Dutch residents using a 36-marker mass cytometry panel. By applying an unbiased multidimensional approach, we observed differences in the balance between the naïve and memory compartments, with higher CD11c+ and double negative (DN-IgDnegCD27neg) memory (M)B-cells in individuals from rural tropical areas, and conversely lower naïve B-cells compared to residents from an area with less pathogen exposure. Furthermore, characterization of total B-cell populations, CD11c+, DN, and Breg cells showed the emergence of specific memory clusters in individuals living in rural tropical areas. Some of these differences were more pronounced in children compared to adults and suggest that a higher microbial exposure accelerates memory B-cell formation, which "normalizes" with age.
Collapse
Affiliation(s)
- Mathilde A M Chayé
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Oscar R J van Hengel
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Astrid L Voskamp
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | | | - Marion H König
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Koen A Stam
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Mikhael D Manurung
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Yoanne D Mouwenda
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
| | - Yvonne A Aryeetey
- Parasitology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Agnes Kurniawan
- Department of Parasitology, Universitas Indonesia, Jakarta, Indonesia
| | - Yvonne C M Kruize
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Erliyani Sartono
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Anne-Marie Buisman
- Laboratory for Immunology of Infectious Diseases and Vaccines, Center for Infectious Diseases Control, National Institute for Public Health and The Environment, Bilthoven, The Netherlands
| | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Tamar Tak
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| | - Hermelijn H Smits
- Leiden University Center for Infectious Diseases (LUCID), LUMC, Leiden, The Netherlands
| |
Collapse
|
7
|
Roozen GVT, Sukwa N, Chirwa M, White JA, Estrada M, Maier N, Turbyfill KR, Laird RM, Suvarnapunya AE, Sayeh A, D’Alessio F, Marion C, Pattacini L, Hoogerwerf MA, Murugan R, Terrinoni M, Holmgren JR, Sirima SB, Houard S, Simuyandi M, Roestenberg M. Safety, Tolerability, and Immunogenicity of the Invaplex AR-DetoxShigella Vaccine Co-Administered with the dmLT Adjuvant in Dutch and Zambian Adults: Study Protocol for a Multi-Center, Randomized, Double-Blind, Placebo-Controlled, Dose-Escalation Phase Ia/b Clinical Trial. Vaccines (Basel) 2025; 13:48. [PMID: 39852827 PMCID: PMC11769217 DOI: 10.3390/vaccines13010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Shigella infections remain endemic in places with poor sanitation and are a leading cause of diarrheal mortality globally, as well as a major contributor to gut enteropathy and stunting. There are currently no licensed vaccines for shigellosis but it has been estimated that an effective vaccine could avert 590,000 deaths over a 20-year period. A challenge to effective Shigella vaccine development has been the low immunogenicity and protective efficacy of candidate Shigella vaccines in infants and young children. Additionally, a new vaccine might be less immunogenic in a highly endemic setting compared to a low endemic setting ("vaccine hyporesponsiveness"). The use of a potent adjuvant enhancing both mucosal and systemic immunity might overcome these problems. InvaplexAR-Detox is an injectable Shigella vaccine that uses a novel combination of conserved invasion plasmid antigen proteins and a serotype-specific bacterial lipopolysaccharide attenuated for safe intramuscular administration. The adjuvant dmLT has been shown to enhance Shigella immune responses in mice, has safely been administered intramuscularly, and was shown to enhance immune responses in healthy volunteers when given in combination with other antigens in phase I trials. This article describes the protocol of a study that will be the first to assess the safety, tolerability, and immunogenicity of InvaplexAR-Detox co-administered with dmLT in healthy adults in low-endemic and high-endemic settings. METHODS In a multi-center, randomized, double-blind, and placebo-controlled dose-escalation phase Ia/b trial, the safety, tolerability, and immunogenicity of three intramuscular vaccinations administered 4 weeks apart with 2.5 µg or 10 µg of InvaplexAR-Detox vaccine, alone or in combination with 0.1 µg of the dmLT adjuvant, will first be assessed in a total of 50 healthy Dutch adults (phase Ia) and subsequently in 35 healthy Zambian adults (phase Ib) aged 18-50 years. The primary outcome is safety, and secondary outcomes are humoral and cellular immune responses to the adjuvanted or non-adjuvanted vaccine. DISCUSSION This trial is part of the ShigaPlexIM project that aims to advance the early clinical development of an injectable Shigella vaccine and to make the vaccine available for late-stage clinical development. This trial addresses the issue of hyporesponsiveness in an early stage of clinical development by testing the vaccine and adjuvant in an endemic setting (Zambia) after the first-in-human administration and the dose-escalation has proven safe and tolerable in a low-endemic setting (Netherlands). Besides strengthening the vaccine pipeline against a major diarrheal disease, another goal of the ShigaPlexIM project is to stimulate capacity building and strengthen global North-South relations in clinical research. TRIAL REGISTRATION EU CT number: 2023-506394-35-02, ClinicalTrials.gov identifier: NCT05961059.
Collapse
Affiliation(s)
- Geert V. T. Roozen
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (L.P.); (R.M.)
| | - Nsofwa Sukwa
- Centre for Infectious Disease Research Zambia, Lusaka P.O. Box 34681, Zambia; (N.S.)
| | - Masuzyo Chirwa
- Centre for Infectious Disease Research Zambia, Lusaka P.O. Box 34681, Zambia; (N.S.)
| | | | | | | | - Kevin R. Turbyfill
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA (A.E.S.)
| | - Renee M. Laird
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA (A.E.S.)
| | | | - Aicha Sayeh
- European Vaccine Initiative, 69115 Heidelberg, Germany (S.H.)
| | | | - Candice Marion
- European Vaccine Initiative, 69115 Heidelberg, Germany (S.H.)
| | - Laura Pattacini
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (L.P.); (R.M.)
| | - Marie-Astrid Hoogerwerf
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (L.P.); (R.M.)
| | - Rajagopal Murugan
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (L.P.); (R.M.)
| | - Manuela Terrinoni
- Department of Microbiology and Immunology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Jan R. Holmgren
- Department of Microbiology and Immunology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Sodiomon B. Sirima
- Groupe de Recherche Action en Santé, Ouagadougou 06 BP 10248, Burkina Faso
| | - Sophie Houard
- European Vaccine Initiative, 69115 Heidelberg, Germany (S.H.)
| | - Michelo Simuyandi
- Centre for Infectious Disease Research Zambia, Lusaka P.O. Box 34681, Zambia; (N.S.)
| | - Meta Roestenberg
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (L.P.); (R.M.)
| |
Collapse
|
8
|
Padilla-Bórquez DL, Matuz-Flores MG, Hernández-Bello J, Rosas-Rodríguez JA, Turrubiates-Hernández FJ, García-Arellano S, González-Estevez G, Ceja-Galvez HR, Oregon-Romero E, López-Reyes A, Muñoz-Valle JF. Influence of previous COVID-19 exposure and vaccine type (CoronaVac, ChAdOx1 nCov-19 or BNT162b2) on antibody and cytokine (Th1 or Th2) responses. Hum Vaccin Immunother 2024; 20:2394265. [PMID: 39246041 PMCID: PMC11385164 DOI: 10.1080/21645515.2024.2394265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024] Open
Abstract
To achieve global herd immunity, widespread vaccination is the most effective strategy. Vaccines stimulate the immune system, generating cytokines and chemokines, isotype antibodies, and neutralizing antibodies; all these molecules collectively provide a more comprehensive characterization of the immune response post-vaccination. We conducted a longitudinal study in northwestern Mexico, involving 120 individuals before vaccination and after the first dose of the SARS-CoV-2 vaccine, and 46 individuals after their second dose. Our findings reveal that antibody levels stabilize over time; cytokine levels generally increase following the first dose but decrease after the second dose and higher than normal levels in IgG1 and IgG3 concentrations are present. Most of the innate cytokines determined in this study were higher after the first dose of the vaccine. Regardless of previous infection history, this finding suggests that the first dose of the vaccine is crucial and may stimulate immunity by enhancing the innate immune response. Conversely, increased levels of IL-4, indicative of a Th2 response, were found in individuals without prior exposure to the virus and in those vaccinated with CoronaVac. These results suggest that the immune response to COVID-19 vaccines is multi-faceted, with preexisting immunity potentiating a more robust innate response. Vaccine type plays a critical role, with genetic vaccines favoring a Th1 response and inactivated vaccines like CoronaVac skewing toward a Th2 profile.
Collapse
Affiliation(s)
- Diana Lourdes Padilla-Bórquez
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Mónica Guadalupe Matuz-Flores
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Jorge Hernández-Bello
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Jesús Alfredo Rosas-Rodríguez
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Navojoa, México
| | - Francisco Javier Turrubiates-Hernández
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Samuel García-Arellano
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Guillermo González-Estevez
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Hazael Ramiro Ceja-Galvez
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Edith Oregon-Romero
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Alberto López-Reyes
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Secretaria de Salud, Ciudad de México, México
| | - Jose Francisco Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| |
Collapse
|
9
|
Cnossen VM, van Leeuwen RP, Mazur NI, Vernhes C, ten Voorde W, Burggraaf J, de Visser SJ, Roestenberg M, Kamerling IMC. From setbacks to success: lessons from the journey of RSV vaccine development. Ther Adv Vaccines Immunother 2024; 12:25151355241308305. [PMID: 39711948 PMCID: PMC11660060 DOI: 10.1177/25151355241308305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024] Open
Abstract
Respiratory syncytial virus (RSV) causes high worldwide infant mortality, as well as a high disease burden in the elderly. Efforts in vaccine development over the past 60 years have recently delivered three approved vaccines and two monoclonal antibodies (mAbs). Looking back at the eventful history of RSV vaccine development, several factors can be identified that have hampered the developmental pathway, including the occurrence of enhanced RSV disease (ERD) in the first vaccine attempt and the difficulty in characterizing and stabilizing the pre-fusion F protein as a vaccine target. Moreover, the need for large trials to test vaccine efficacy, usually done late in development, and the lack of a correlate of protection (CoP) result in significant uncertainties in RSV vaccine development. The use of controlled human infection models (CHIMs) may provide a solution for some of these problems: through swift, cost-efficient and closely monitored assessment of vaccine safety and efficacy in early clinical phases, vaccines can either 'fail fast' or show results supporting further investments. Moreover, CHIMs facilitate the assessment of disease and could assist in the identification of a CoP supporting late-stage development. Although some factors may affect translatability to real-world vaccine efficacy, CHIMs can support the clinical development pathway in various ways. We advocate for, and demonstrate, a conceptual and rational design of RSV vaccine development. Assessing protective efficacy early on would result in the most cost-efficient pathway and identification of target populations should be done as early as possible. For RSV, elderly individuals and people in low- and middle-income countries are high-impact populations for RSV prevention. While RSV immunization is now available in certain regions, global access is not accomplished yet, and worldwide prevention does not seem within reach. Quick and cost-effective assessments of candidates currently in the pipeline could contribute to future successes in the battle against RSV.
Collapse
Affiliation(s)
- Victor M. Cnossen
- Centre for Human Drug Research, Zernikedreef 8, 2333 CL Leiden, The Netherlands
| | | | | | - Charlotte Vernhes
- Vaccines Europe, European Federation of Pharmaceutical Industries and Associations, Brussels, Belgium
| | | | | | - Saco J. de Visser
- Centre for Future Affordable & Sustainable Therapy Development (FAST), The Hague, The Netherlands
| | - Meta Roestenberg
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands
| | - Ingrid M. C. Kamerling
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
10
|
Nassuuna J, Zirimenya L, Nkurunungi G, Natukunda A, Zziwa C, Ninsiima C, Apule B, Onen C, Amongi S, Serubanja J, Tumwesige P, Nsubuga D, Amongin R, van Dam GJ, Corstjens PLAM, Kayiwa J, Kabagenyi J, Cose S, Wajja A, Kaleebu P, Webb EL, Elliott AM. The effect of BCG revaccination on the response to unrelated vaccines in urban Ugandan adolescents (POPVAC C): an open-label, randomised controlled trial. Lancet Glob Health 2024; 12:e1849-e1859. [PMID: 39424573 PMCID: PMC11483248 DOI: 10.1016/s2214-109x(24)00282-1] [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: 12/15/2023] [Revised: 05/10/2024] [Accepted: 06/27/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Immune responses induced by several important vaccines differ between populations, with reduced responses in low-income and rural settings compared with high-income and urban settings. BCG immunisation boosts immune responses to some unrelated vaccines in high-income populations. We aimed to test the hypothesis that BCG revaccination can enhance responses to unrelated vaccines in Ugandan schoolchildren. METHODS We conducted an open-label, randomised controlled trial to compare the effects of BCG revaccination versus no BCG revaccination on the immunogenicity of subsequent unrelated vaccines among adolescents aged 13-17 years who are participants in an urban Ugandan birth cohort study, in which BCG vaccination was documented at birth. Participants were excluded if they had received any of the trial vaccines or related agents when aged 5 years or older. Computer-generated 1:1 randomisation was implemented in REDCap. Participants were excluded if they were concurrently enrolled in other trials; had a clinically significant history of immunodeficiency, or serious psychiatric conditions or moderate to severe acute illnesses; were taking immunosuppressive medications; had allergies to vaccine components, a predisposition towards developing keloid scarring; positive HIV tests or pregnancy tests; were female participants who were lactating; or if they planned to use investigational drugs, vaccines, blood products, or any combination thereof. Trial participants assigned to the BCG revaccination group received the live parenteral BCG-Russia vaccine (Serum Institute of India, Pune, India; 0·1 mL intradermally, right upper arm) at week 0. All participants received yellow fever vaccine (YF-17D; Sanofi Pasteur, Lyon, France; 0·5 mL intramuscularly, left upper arm), live oral typhoid vaccine (Ty21a; PaxVax, London, UK; one capsule per day taken for three alternate days), and quadrivalent virus-like particle human papillomavirus (HPV) vaccine (Merck, Rahway, NJ, USA; 0·5 mL intramuscularly, left upper arm) at week 4; and toxoid vaccines (tetanus-diphtheria; Serum Institute of India; 0·5 mL intramuscularly, left upper arm) and an HPV booster at week 28. An additional HPV vaccination at week 8 was provided to female participants older than 14 years who had not previously been vaccinated. The primary outcomes were yellow fever neutralising antibody titres at 4 weeks post-YF-17D vaccination, Salmonella enterica serovar Typhi (henceforth S Typhi) O-lipopolysaccharide (O:LPS)-specific IgG concentration at 4 weeks post-Ty21a vaccination, and HPV-16 and HPV-18 L1 protein-specific IgG concentration at 4 weeks post-HPV vaccination. Primary outcome assays were conducted at week 8, and at week 52 for tetanus-diphtheria. We conducted an intention-to-treat analysis comparing log-transformed outcomes between trial groups, with results back-transformed to geometric mean ratios (GMRs). The safety population comprised all randomly allocated participants. The trial was registered at the ISRCTN Registry (ISRCTN10482904) and is complete. FINDINGS Between Aug 31 and Oct 12, 2020, we screened 376 potential participants for eligibility. We enrolled and randomly allocated 300 participants to the two groups (151 [50%] to the BCG group and 149 [50%] to the no BCG group). 178 (59%) of 300 participants were male and 122 (41%) were female. 142 (91%) of 151 participants in the BCG group and 139 (93%) of 149 in the no BCG group completed follow-up. There was no effect of BCG revaccination, compared with no BCG revaccination, on the response observed for any vaccine. Yellow fever plaque reduction neutralising reference tests (PRNT50) titres (the reciprocal of the last plasma dilution that reduced by 50%) had a GMR of 0·95 (95% CI 0·75-1·19; p=0·62) and PRNT90 (reciprocal of the last plasma dilution that reduced by 90%) had a GMR of 0·94 (0·74-1·19; p=0·60); IgG to S Typhi O:LPS was 0·99 (0·80-1·23; p=0·94); IgG to HPV-16 was 0·97 (0·69-1·35; p=0·85) and to HPV-18 was 1·03 (0·76-1·40; p=0·83); and toxoid-specific IgG for tetanus was 1·13 (0·87-1·47; p=0·36) and was 1·00 (0·87-1·16; p=0·97) for diphtheria. There were no serious adverse events in either group. INTERPRETATION We found no evidence that BCG revaccination is an effective strategy to improve immunogenicity of other vaccines in this low-income, urban setting. FUNDING UK Medical Research Council. TRANSLATION For the Luganda translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Jacent Nassuuna
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Ludoviko Zirimenya
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda; Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | - Gyaviira Nkurunungi
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda; Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK.
| | - Agnes Natukunda
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda; International Statistics and Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Christopher Zziwa
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Caroline Ninsiima
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Barbara Apule
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Caroline Onen
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Susan Amongi
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Joel Serubanja
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Pius Tumwesige
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Denis Nsubuga
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Rebecca Amongin
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Govert J van Dam
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Paul L A M Corstjens
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - John Kayiwa
- Department of Arbovirology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Joyce Kabagenyi
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Stephen Cose
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda; Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | - Anne Wajja
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda; Department of Global Health and Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Pontiano Kaleebu
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Emily L Webb
- International Statistics and Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Alison M Elliott
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda; Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
11
|
Nono JK. Intensify praziquantel administration to reverse vaccine hyporesponsiveness in LMICs? Lancet Glob Health 2024; 12:e1746-e1747. [PMID: 39424565 DOI: 10.1016/s2214-109x(24)00356-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 10/21/2024]
Affiliation(s)
- Justin Komguep Nono
- Unit of Immunobiology and Helminth Infections, Laboratory of Molecular Biology and Biotechnology, Institute of Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon; Division of Immunology, Health Science Faculty, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
12
|
Pyuza JJ, van Dorst MM, Stam K, Wammes L, König M, Kullaya VI, Kruize Y, Huisman W, Andongolile N, Ngowi A, Shao ER, Mremi A, Hogendoorn PC, Msuya SE, Jochems SP, de Steenhuijsen Piters WA, Yazdanbakhsh M. Lifestyle score is associated with cellular immune profiles in healthy Tanzanian adults. Brain Behav Immun Health 2024; 41:100863. [PMID: 39398291 PMCID: PMC11470418 DOI: 10.1016/j.bbih.2024.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/31/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Immune system and vaccine responses vary across geographical locations worldwide, not only between high and low-middle income countries (LMICs), but also between rural and urban populations within the same country. Lifestyle factors such as housing conditions, exposure to microorganisms and parasites and diet are associated with rural-and urban-living. However, the relationships between these lifestyle factors and immune profiles have not been mapped in detail. Here, we profiled the immune system of 100 healthy Tanzanians living across four rural/urban areas using mass cytometry. We developed a lifestyle score based on an individual's household assets, housing condition and recent dietary history and studied the association with cellular immune profiles. Seventeen out of 80 immune cell clusters were associated with living location or lifestyle score, with eight identifiable only using lifestyle score. Individuals with low lifestyle score, most of whom live in rural settings, showed higher frequencies of NK cells, plasmablasts, atypical memory B cells, T helper 2 cells, regulatory T cells and activated CD4+ T effector memory cells expressing CD38, HLA-DR and CTLA-4. In contrast, those with high lifestyle score, most of whom live in urban areas, showed a less activated state of the immune system illustrated by higher frequencies of naïve CD8+ T cells. Using an elastic net machine learning model, we identified cellular immune signatures most associated with lifestyle score. Assuming a link between these immune profiles and vaccine responses, these signatures may inform us on the cellular mechanisms underlying poor responses to vaccines, but also reduced autoimmunity and allergies in low- and middle-income countries.
Collapse
Affiliation(s)
- Jeremia J. Pyuza
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Institute of Public Health, Kilimanjaro Christian University Medical College (KCMUCo), Moshi, Tanzania
- Kilimanjaro Clinical Research Institute (KCRI), Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Marloes M.A.R. van Dorst
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Koen Stam
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Linda Wammes
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Marion König
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Vesla I. Kullaya
- Kilimanjaro Clinical Research Institute (KCRI), Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical College (KCMUCo), Moshi, Tanzania
| | - Yvonne Kruize
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Wesley Huisman
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Nikuntufya Andongolile
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Anastazia Ngowi
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Elichilia R. Shao
- Department of Internal Medicine, Kilimanjaro Christian Medical University College (KCMUCo), Moshi, Tanzania
- Department of Internal Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Alex Mremi
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | | | - Sia E. Msuya
- Institute of Public Health, Kilimanjaro Christian University Medical College (KCMUCo), Moshi, Tanzania
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Simon P. Jochems
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | | | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| |
Collapse
|
13
|
Mukhiya R, Fleischmann WA, Loughland JR, Chan JA, de Labastida Rivera F, Andrew D, Beeson JG, McCarthy JS, Barber BE, Lopez JA, Engwerda C, Thomson-Luque R, Boyle MJ. Heterogeneity of the human immune response to malaria infection and vaccination driven by latent cytomegalovirus infection. EBioMedicine 2024; 109:105419. [PMID: 39490199 PMCID: PMC11576503 DOI: 10.1016/j.ebiom.2024.105419] [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: 08/05/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Human immune responses to infection and vaccination are heterogenous, driven by multiple factors including genetics, environmental exposures and personal infection histories. For malaria caused by Plasmodium falciparum parasites, host factors that impact on humoral immunity are poorly understood. METHODS We investigated the role of latent cytomegalovirus (CMV) on the host immune response to malaria using samples obtained from individuals in previously conducted Phase 1 trials of blood stage P. falciparum Controlled Human Malaria Infection (CHMI) and in a MSP1 vaccine clinical trial. Induced antibody and functions of antibodies, as well as CD4 T cell responses were quantified. FINDINGS CMV seropositivity was associated with reduced induction of parasite specific antibodies following malaria infection and vaccination. During infection, reduced antibody induction was associated with modifications to the T -follicular helper (Tfh) cell compartment. CMV seropositivity was associated with a skew towards Tfh1 cell subsets before and after malaria infection, and reduced activation of Tfh2 cells. Protective Tfh2 cell activation was only associated with antibody development in individuals who were CMV seronegative, and a higher proportion of Tfh1 cells was associated with lower antibody development in individuals who were CMV seropositive. During MSP1 vaccination, reduced antibody induction in individuals who were CMV seropositive was associated with CD4 T cell expression of terminal differentiation marker CD57. INTERPRETATION These findings suggest that CMV seropositivity may be negatively associated with malaria antibody development. Further studies in larger cohorts, particularly in malaria endemic regions are required to investigate whether CMV infection may modify immunity to malaria gained during infection or vaccination in children. FUNDING Work was funded by National Health and Medical Research Council of Australia, CSL Australia and Snow Medical Foundation. Funders had no role in data generation, writing of manuscript of decision to submit for publication.
Collapse
Affiliation(s)
- Reena Mukhiya
- Burnet Institute, Melbourne, Australia; School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Wim A Fleischmann
- Center for Infectious Diseases, Virology, Heidelberg University, Medical Faculty, University Hospital Heidelberg, Germany
| | - Jessica R Loughland
- Burnet Institute, Melbourne, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jo-Anne Chan
- Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, University of Melbourne, Australia; Department of Microbiology and School of Translational Medicine, Monash University, Australia
| | | | - Dean Andrew
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - James G Beeson
- Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, University of Melbourne, Australia; Department of Microbiology and School of Translational Medicine, Monash University, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia; Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | | | - J Alejandro Lopez
- School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Christian Engwerda
- School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Richard Thomson-Luque
- Sumaya-Biotech GmbH & Co. KG, Germany; Centre for Infectious Diseases, Parasitology, Heidelberg University, Medical Faculty, University Hospital Heidelberg, Germany
| | - Michelle J Boyle
- Burnet Institute, Melbourne, Australia; School of Environmental Sciences, Griffith University, Brisbane, Australia; QIMR Berghofer Medical Research Institute, Brisbane, Australia; Department of Infectious Diseases, University of Melbourne, Australia; Department of Microbiology and School of Translational Medicine, Monash University, Australia.
| |
Collapse
|
14
|
Zirimenya L, Natukunda A, Nassuuna J, Kabagenyi J, Nkurunungi G, Elliott AM, Webb EL. The Effect of Malaria on Responses to Unrelated Vaccines in Animals and Humans: A Systematic Review and Meta-Analysis. Parasite Immunol 2024; 46:e13067. [PMID: 39439428 DOI: 10.1111/pim.13067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/21/2024] [Indexed: 10/25/2024]
Abstract
Vaccine efficacy varies globally, often showing reduced immune responses in low- and middle-income countries, possibly due to the immunomodulatory effects of parasitic infections like malaria. This systematic review evaluates the impact of malaria on immune responses to unrelated vaccines in humans and animals. We systematically searched five databases-MEDLINE, Web of Science, Global Health, Scopus and Embase-up to 5th December 2023. Eligible studies compared immune responses to WHO-approved vaccines between malaria-infected and uninfected groups, or between antimalarial-treated and untreated groups. Meta-analysis was performed using random-effects models with standardised mean differences (SMDs) as summary statistics. The study is registered with PROSPERO (CRD42022298053). Twenty-four articles (17 human, 7 animal) met the inclusion criteria, with 13 human articles contributing data for the meta-analysis. Significant heterogeneity was observed. Vaccine responses were higher in malaria uninfected individuals (SMD 0.34, 95% CI 0.07 to 0.60, I2 = 87.15%) with weaker differences between antimalarial-treated and untreated groups (SMD 0.07, 95% CI -0.01 to 0.16, I2 = 85.01%). The overall SMD for malaria uninfected/treated vs. infected/untreated was 0.15, 95% CI 0.05-0.26, I2 = 90.91. Narrative analysis suggested malaria's adverse impact on vaccine responses in animals. Malaria infection may impair vaccines responses; with preventive treatment of malaria partially reversing these effects, highlighting the need for targeted public health interventions.
Collapse
Affiliation(s)
- Ludoviko Zirimenya
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine (MRC/UVRI and LSHTM) Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Agnes Natukunda
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine (MRC/UVRI and LSHTM) Uganda Research Unit, Entebbe, Uganda
- International Statistics and Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Jacent Nassuuna
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine (MRC/UVRI and LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Joyce Kabagenyi
- School of Infection and Immunity, University of Glasgow, Glasgow, Scotland
| | - Gyaviira Nkurunungi
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine (MRC/UVRI and LSHTM) Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Alison M Elliott
- Immunomodulation and Vaccines Focus Area, Vaccine Research Theme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine (MRC/UVRI and LSHTM) Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Emily L Webb
- International Statistics and Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
15
|
Abavisani M, Ansari B, Ebadpour N, Sahebkar A. How does geographical diversity shape vaccine efficacy? Clin Exp Vaccine Res 2024; 13:271-300. [PMID: 39525670 PMCID: PMC11543789 DOI: 10.7774/cevr.2024.13.4.271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/22/2024] [Accepted: 06/04/2024] [Indexed: 11/16/2024] Open
Abstract
Vaccination is a cornerstone of public health, saving millions of lives each year by preventing a variety of infectious diseases. Yet, despite global vaccination efforts, emerging research highlights significant geographical disparities in vaccine efficacy and immunogenicity. These variations underscore the critical interplay between immunological factors and environmental, genetic, and nutritional elements across different populations. Our review article aimed to explore the multifactorial reasons behind geographical variations in vaccine efficacy. Also, this study has shown how important host factors like age, obesity, gender, and genetic diversity, especially within the major histocompatibility complex, are in determining how well a vaccine works. Nutritional status, namely deficiencies in micronutrients such as vitamins and zinc, and lifestyle factors including stress, sleep, alcohol consumption, and physical activity are also shown to have profound effects on vaccine-induced immunity. Importantly, our paper also brought to light the influence of microbial and ecological factors, such as the gut microbiome and environmental pollutants, on the immune system's response to vaccination. The findings emphasize the importance of tailoring vaccination strategies to accommodate the unique immunological landscapes shaped by geographical and societal factors. This tailored approach could enhance vaccine efficacy, reduce disparities in vaccine response, and ultimately contribute to the global fight against infectious diseases.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahareh Ansari
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Boyle MJ, Engwerda CR, Jagannathan P. The impact of Plasmodium-driven immunoregulatory networks on immunity to malaria. Nat Rev Immunol 2024; 24:637-653. [PMID: 38862638 PMCID: PMC11688169 DOI: 10.1038/s41577-024-01041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 06/13/2024]
Abstract
Malaria, caused by infection with Plasmodium parasites, drives multiple regulatory responses across the immune landscape. These regulatory responses help to protect against inflammatory disease but may in some situations hamper the acquisition of adaptive immune responses that clear parasites. In addition, the regulatory responses that occur during Plasmodium infection may negatively affect malaria vaccine efficacy in the most at-risk populations. Here, we discuss the specific cellular mechanisms of immunoregulatory networks that develop during malaria, with a focus on knowledge gained from human studies and studies that involve the main malaria parasite to affect humans, Plasmodium falciparum. Leveraging this knowledge may lead to the development of new therapeutic approaches to increase protective immunity to malaria during infection or after vaccination.
Collapse
Affiliation(s)
- Michelle J Boyle
- Life Sciences Division, Burnet Institute, Melbourne, Victoria, Australia.
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| | | | - Prasanna Jagannathan
- Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
17
|
Ongwe MEB, Mouwenda YD, Manurung MD, Heieis G, Azimi S, Adegnika AA, Kremsner PG, Kuijpers TW, Yazdanbakhsh M, Everts B. Potentiation of the axis involving pentose phosphate pathway/NADPH oxidase/reactive oxygen species drives higher IL-10 production in monocytes of Sub-Saharan Africans. Eur J Immunol 2024; 54:e2451029. [PMID: 38873882 DOI: 10.1002/eji.202451029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
Cellular metabolism is a key determinant of immune cell function. Here we found that CD14+ monocytes from Sub-Saharan Africans produce higher levels of IL-10 following TLR-4 stimulation and are bioenergetically distinct from monocytes from Europeans. Through metabolomic profiling, we identified the higher IL-10 production to be driven by increased baseline production of NADPH oxidase-dependent reactive oxygen species, supported by enhanced pentose phosphate pathway activity. Together, these data indicate that NADPH oxidase-derived ROS is a metabolic checkpoint in monocytes that governs their inflammatory profile and uncovers a metabolic basis for immunological differences across geographically distinct populations.
Collapse
Affiliation(s)
- Madeleine Eunice Betouke Ongwe
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut de Recherches en Écologie Tropicale, Centre National de la Recherche Scientifique et Technologique, Libreville, Gabon
| | - Yoanne D Mouwenda
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Mikhael D Manurung
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Graham Heieis
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Shohreh Azimi
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Ayola A Adegnika
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
- German Center for Infection Research, Tübingen, Germany
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Taco W Kuijpers
- Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Emma Children's Hospital, Academic Medical Center, Dept of Paediatric Immunology, Rheumatology and Infectious Diseases, University of Amsterdam, Amsterdam, the Netherlands
| | - Maria Yazdanbakhsh
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Bart Everts
- Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
18
|
Liu X, Xiao H, Cui P, Chen J, Chao J, Wu X, Lu J, Zhang X, Xu G, Liu Y. Differential polyvalent passive immune protection of egg yolk antibodies (IgY) against live and inactivated Vibrio fluvialis in fish. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109751. [PMID: 38971349 DOI: 10.1016/j.fsi.2024.109751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Egg yolk antibodies (IgY) can be prepared in large quantities and economically, and have potential value as polyvalent passive vaccines (against multiple bacteria) in aquaculture. This study prepared live and inactivated Vibrio fluvialis IgY and immunized Carassius auratus prior to infection with V. fluvialis and Aeromonas hydrophila. The results showed that the two IgY antibodies hold effective passive protective rates against V. fluvialis and A. hydrophila in C. auratus. Further, the serum of C. auratus recognized the two bacteria in vitro, with a decrease in the bacteria content of the kidney. The phagocytic activity of C. auratus plasma was enhanced, with a decrease in the expression of inflammatory and antioxidant factors. Pathological sections showed that the kidney, spleen, and intestinal tissue structures were intact, and apoptosis and DNA damage decreased in kidney cells. Moreover, the immunoprotection conferred by the live V. fluvialis IgY was higher than that of the inactivated IgY. Addition, live V. fluvialis immunity induced IgY antibodies against outer membrane proteins of V. fluvialis were more than inactivated V. fluvialis immunity. Furthermore, heterologous immune bacteria will not cause infection, so V. fluvialis can be used to immunize chickens to obtain a large amount of IgY antibody. These findings suggest that the passive immunization effect of live bacterial IgY antibody on fish is significantly better than that of inactivated bacterial antibody, and the live V. fluvialis IgY hold potential value as polyvalent passive vaccines in aquaculture.
Collapse
Affiliation(s)
- Xiang Liu
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, Fuyang Normal University, Fuyang, 236041, China; Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China; Chinese-German Joint Institute for Natural Product Research, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, 723000, China.
| | - Huihui Xiao
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, Fuyang Normal University, Fuyang, 236041, China; Fuyang Normal University--Funan Rural Revitalization Collaborative Technology Service Center, Fuyang Normal University, Fuyang, 236041, China
| | - Pan Cui
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, Fuyang Normal University, Fuyang, 236041, China
| | - Jing Chen
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, Fuyang Normal University, Fuyang, 236041, China
| | - Jia Chao
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, Fuyang Normal University, Fuyang, 236041, China; Chinese-German Joint Institute for Natural Product Research, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, 723000, China
| | - Xiaoqing Wu
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, Fuyang Normal University, Fuyang, 236041, China
| | - Juan Lu
- Fuyang Normal University--Funan Rural Revitalization Collaborative Technology Service Center, Fuyang Normal University, Fuyang, 236041, China
| | - Xiaoying Zhang
- Chinese-German Joint Institute for Natural Product Research, College of Biological Science and Engineering, Shaanxi University of Technology, Hanzhong, 723000, China
| | - Gaoxiao Xu
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, Fuyang Normal University, Fuyang, 236041, China.
| | - Yong Liu
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, Fuyang Normal University, Fuyang, 236041, China; Fuyang Normal University--Funan Rural Revitalization Collaborative Technology Service Center, Fuyang Normal University, Fuyang, 236041, China.
| |
Collapse
|
19
|
Miles JR, Lu P, Bai S, Aguillón-Durán GP, Rodríguez-Herrera JE, Gunn BM, Restrepo BI, Lu LL. Antigen specificity shapes antibody functions in tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597169. [PMID: 38895452 PMCID: PMC11185737 DOI: 10.1101/2024.06.03.597169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Tuberculosis (TB) is the number one infectious disease cause of death worldwide due to an incomplete understanding of immunity. Emerging data highlight antibody functions mediated by the Fc domain as immune correlates. However, the mechanisms by which antibody functions impact the causative agent Mycobacterium tuberculosis (Mtb) are unclear. Here, we examine how antigen specificity determined by the Fab domain shapes Fc effector functions against Mtb. Using the critical structural and secreted virulence proteins Mtb cell wall and ESAT-6 & CFP-10, we observe that antigen specificity alters subclass, antibody post-translational glycosylation, and Fc effector functions in TB patients. Moreover, Mtb cell wall IgG3 enhances disease through opsonophagocytosis of extracellular Mtb . In contrast, polyclonal and a human monoclonal IgG1 we generated targeting ESAT-6 & CFP-10 inhibit intracellular Mtb . These data show that antibodies have multiple roles in TB and antigen specificity is a critical determinant of the protective and pathogenic capacity.
Collapse
|
20
|
Animal research is not always king: researchers should explore the alternatives. Nature 2024; 631:481. [PMID: 39014040 DOI: 10.1038/d41586-024-02314-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
|
21
|
Zhu F, Zheng W, Gong Y, Zhang J, Yu Y, Zhang J, Liu M, Guan F, Lei J. Trichinella spiralis Infection Inhibits the Efficacy of RBD Protein of SARS-CoV-2 Vaccination via Regulating Humoral and Cellular Immunity. Vaccines (Basel) 2024; 12:729. [PMID: 39066367 PMCID: PMC11281533 DOI: 10.3390/vaccines12070729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/23/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Vaccines are the most effective and feasible way to control pathogen infection. Helminths have been reported to jeopardize the protective immunity mounted by several vaccines. However, there are no experimental data about the effect of helminth infection on the effectiveness of COVID-19 vaccines. Here, a mouse model of trichinosis, a common zoonotic disease worldwide, was used to investigate effects of Trichinella spiralis infection on the RBD protein vaccine of SARS-CoV-2 and the related immunological mechanism, as well as the impact of albendazole (ALB) deworming on the inhibitory effect of the parasite on the vaccination. The results indicated that both the enteric and muscular stages of T. spiralis infection inhibited the vaccine efficacy, evidenced by decreased levels of IgG, IgM, sIgA, and reduced serum neutralizing antibodies, along with suppressed splenic germinal center (GC) B cells in the vaccinated mice. Pre-exposure to trichinosis promoted Th2 and/or Treg immune responses in the immunized mice. Furthermore, ALB treatment could partially reverse the inhibitory effect of T. spiralis infection on the efficiency of the vaccination, accompanied by a restored proportion of splenic GC B cells. Therefore, given the widespread prevalence of helminth infections worldwide, deworming therapy needs to be considered when implementing COVID-19 vaccination strategies.
Collapse
Affiliation(s)
- Feifan Zhu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; (F.Z.); (W.Z.); (Y.G.); (J.Z.)
| | - Wenwen Zheng
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; (F.Z.); (W.Z.); (Y.G.); (J.Z.)
| | - Yiyan Gong
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; (F.Z.); (W.Z.); (Y.G.); (J.Z.)
| | - Jinyuan Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; (F.Z.); (W.Z.); (Y.G.); (J.Z.)
| | - Yihan Yu
- Department of Pulmonary Medicine, Hubei Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan 430015, China; (Y.Y.); (J.Z.); (M.L.)
| | - Jixian Zhang
- Department of Pulmonary Medicine, Hubei Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan 430015, China; (Y.Y.); (J.Z.); (M.L.)
| | - Mengjun Liu
- Department of Pulmonary Medicine, Hubei Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan 430015, China; (Y.Y.); (J.Z.); (M.L.)
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; (F.Z.); (W.Z.); (Y.G.); (J.Z.)
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China; (F.Z.); (W.Z.); (Y.G.); (J.Z.)
| |
Collapse
|
22
|
Doshi RH, Mukadi PK, Casey RM, Kizito GM, Gao H, Nguete U B, Laven J, Sabi L, Kaba DK, Muyembe-Tamfum JJ, Hyde TB, Ahuka-Mundeke S, Staples JE. Immunological response to fractional-dose yellow fever vaccine administered during an outbreak in Kinshasa, Democratic Republic of the Congo: results 5 years after vaccination from a prospective cohort study. THE LANCET. INFECTIOUS DISEASES 2024; 24:611-618. [PMID: 38335976 PMCID: PMC11542170 DOI: 10.1016/s1473-3099(23)00809-5] [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: 10/24/2023] [Revised: 12/03/2023] [Accepted: 12/14/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND In 2016, outbreaks of yellow fever in Angola and the Democratic Republic of the Congo led to a global vaccine shortage. A fractional dose of 17DD yellow fever vaccine (containing one-fifth [0·1 ml] of the standard dose) was used during a pre-emptive mass campaign in August, 2016, in Kinshasa, Democratic Republic of the Congo among children aged 2 years and older and non-pregnant adults (ie, those aged 18 years and older). 1 year following vaccination, 97% of participants were seropositive; however, the long-term durability of the immune response is unknown. We aimed to conduct a prospective cohort study and invited participants enrolled in the previous evaluation to return 5 years after vaccination to assess durability of the immune response. METHODS Participants returned to one of six health facilities in Kinshasa in 2021, where study staff collected a brief medical history and blood specimen. We assessed neutralising antibody titres against yellow fever virus using a plaque reduction neutralisation test with a 50% cutoff (PRNT50). Participants with a PRNT50 titre of 10 or higher were considered seropositive. The primary outcome was the proportion of participants seropositive at 5 years. FINDINGS Among the 764 participants enrolled, 566 (74%) completed the 5-year visit. 5 years after vaccination, 539 (95·2%, 95% CI 93·2-96·7) participants were seropositive, including 361 (94·3%, 91·5-96·2) of 383 who were seronegative and 178 (97·3%, 93·8-98·8) of 183 who were seropositive at baseline. Geometric mean titres (GMTs) differed significantly across age groups for those who were initially seronegative with the lowest GMT among those aged 2-5 years and highest among those aged 13 years and older. INTERPRETATION A fractional dose of the 17DD yellow fever vaccine induced an immunologic response with detectable titres at 5 years among the majority of participants in the Democratic Republic of the Congo. These findings support the use of fractional-dose vaccination for outbreak prevention with the potential for sustained immunity. FUNDING Gavi, the Vaccine Alliance through the CDC Foundation. TRANSLATION For the French translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Reena H Doshi
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Patrick K Mukadi
- Centers for Disease Control and Prevention Foundation, Atlanta, GA, USA; Department of Clinical Tropical Medicine, Institute of Tropical Medicine, Nagasaki University Graduate School of Biomedical Science, Nagasaki, Japan
| | - Rebecca M Casey
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Gabriel M Kizito
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Hongjiang Gao
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Beatrice Nguete U
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo
| | - Janeen Laven
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, USA
| | - Lilliane Sabi
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Didine K Kaba
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo
| | | | - Terri B Hyde
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Steve Ahuka-Mundeke
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - J Erin Staples
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, USA
| |
Collapse
|
23
|
Muwanda F, Sendagire H, Mboowa G, Kateete DP, Achan B, Mupere E, Kafeero HM, Bagaya BS. A systematic review reveals that African children of 15-17 years demonstrate low hepatitis B vaccine seroprotection rates. Sci Rep 2023; 13:22182. [PMID: 38092870 PMCID: PMC10719251 DOI: 10.1038/s41598-023-49674-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 12/11/2023] [Indexed: 12/17/2023] Open
Abstract
Childhood HBV immunization remains globally fundamental to the elimination of hepatitis B virus (HBV). However, monitoring proportions of HBV vaccine seroprotection and their determinants among African Pediatric recipients is crucial. This study sought to verify extent of immune protection accorded by the HBV vaccine in African children of up to 17 years of age by pooling the prevalence of seroprotection reported by primary studies conducted in the Northern, Western, and Southern African regions. We included 19 eligible articles out of the 197 initially downloaded, published from 1999 to 2021 from African Journals Online (AJOL), EMBASE, Scopus, and PubMed. The study protocol was registered with the International Prospective Register of Systematic Reviews (PROSPERO), University of York Centre for Reviews and Dissemination, under the registration number CRD42022361277. Significantly higher (p < 0.0001) proportion of HBV vaccine seroprotection (69.07%) was found among children under 15 years of age than children 15-17 years (32.368%), 95% CI [34.2454-39.0847%]. Whereas successful integration of the HBV vaccine on the extended programs on immunizations (EPI) has been a major achievement in the reduction of HBV infection in Africa, markedly reduced HBV vaccine seroprotection is persistently demonstrated among adolescent children 15-17 years of age. Future studies are required to clarify the need for booster dose vaccination in most at risk populations and age groups.
Collapse
Affiliation(s)
- Fahad Muwanda
- Department of Medical Microbiology, Faculty of Health Sciences, Habib Medical School, Islamic University in Uganda, P.O. Box 7689, Kampala, Uganda.
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda.
| | - Hakim Sendagire
- Department of Medical Microbiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Gerald Mboowa
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
- The African Center of Excellence in Bioinformatics and Data-Intensive Sciences, Infectious Diseases Institute, College of Health Sciences, Makerere University, P.O. Box 22418, Kampala, Uganda
| | - David Patrick Kateete
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Beatrice Achan
- Department of Medical Microbiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Ezekiel Mupere
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Hussein Mukasa Kafeero
- Department of Medical Microbiology, Faculty of Health Sciences, Habib Medical School, Islamic University in Uganda, P.O. Box 7689, Kampala, Uganda
- Department of Medical Microbiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Bernard Ssentalo Bagaya
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
| |
Collapse
|