1
|
Avolio E, Bassani B, Campanile M, Mohammed KA, Muti P, Bruno A, Spinetti G, Madeddu P. Shared molecular, cellular, and environmental hallmarks in cardiovascular disease and cancer: Any place for drug repurposing? Pharmacol Rev 2025; 77:100033. [PMID: 40148035 DOI: 10.1016/j.pharmr.2024.100033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer and cardiovascular disease (CVD) are the 2 biggest killers worldwide. Specific treatments have been developed for the 2 diseases. However, mutual therapeutic targets should be considered because of the overlap of cellular and molecular mechanisms. Cancer research has grown at a fast pace, leading to an increasing number of new mechanistic treatments. Some of these drugs could prove useful for treating CVD, which realizes the concept of cancer drug repurposing. This review provides a comprehensive outline of the shared hallmarks of cancer and CVD, primarily ischemic heart disease and heart failure. We focus on chronic inflammation, altered immune response, stromal and vascular cell activation, and underlying signaling pathways causing pathological tissue remodeling. There is an obvious scope for targeting those shared mechanisms, thereby achieving reciprocal preventive and therapeutic benefits. Major attention is devoted to illustrating the logic, advantages, challenges, and viable examples of drug repurposing and discussing the potential influence of sex, gender, age, and ethnicity in realizing this approach. Artificial intelligence will help to refine the personalized application of drug repurposing for patients with CVD. SIGNIFICANCE STATEMENT: Cancer and cardiovascular disease (CVD), the 2 biggest killers worldwide, share several underlying cellular and molecular mechanisms. So far, specific therapies have been developed to tackle the 2 diseases. However, the development of new cardiovascular drugs has been slow compared with cancer drugs. Understanding the intersection between pathological mechanisms of the 2 diseases provides the basis for repurposing cancer therapeutics for CVD treatment. This approach could allow the rapid development of new drugs for patients with CVDs.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy
| | - Marzia Campanile
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Khaled Ak Mohammed
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom; Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paola Muti
- IRCCS MultiMedica, Milan, Italy; Department of Biomedical, Surgical and Dental Health Sciences, University of Milan, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy; Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Paolo Madeddu
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| |
Collapse
|
2
|
Vashi B, Pettrone K, Wilson CS, Chenoweth JG, Brandsma J, Gregory MK, Genzor P, Striegel DA, Rothman RE, Hansoti B, Avornu GD, McBryde B, Zimmerman LR, Woods CW, Petzold EA, Cowden J, Nitayaphan S, Nasomsong W, Clark DV. COVID-19 symptom severity and duration among outpatients, July 2021-May 2023: The PROTECT observational study. PLoS One 2025; 20:e0314518. [PMID: 39982872 PMCID: PMC11844841 DOI: 10.1371/journal.pone.0314518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/11/2024] [Indexed: 02/23/2025] Open
Abstract
INTRODUCTION With the emergence of new SARS-CoV-2 variants has come significant variations in disease manifestation, severity, and duration in non-hospitalized infected patients. To characterize symptom patterns and risk factors associated with symptom severity and duration, COVID-19 and influenza-like illness (ILI) outpatients and their contacts were enrolled at two sites in the United States of America and one site in Thailand. METHODS COVID-19 infection was confirmed at enrollment with a positive antigen or PCR test. Baseline demographics and medical histories were collected from participants at enrollment and daily self-reported symptom questionnaires were obtained to assess symptom severity and duration. Risk factors associated with symptom severity and duration were determined by multivariate logistic regression and Cox proportional hazards model. RESULTS Two hundred and forty one participants meeting the eligibility criteria were enrolled, including 174 confirmed COVID-19 cases (9% Delta and 90% Omicron), 33 ILI cases, and 34 healthy contacts. COVID-19 participants had a shorter median symptom duration of 9.0 (95% CI, 8.0-11.0) days than ILI participants. Infection with the Delta variant resulted in a longer symptom alleviation period compared to infection with the Omicron variant. The most commonly reported symptoms among COVID-19 participants were reported in the nasal and chest/respiratory domains of the FLU-PRO Plus. Participants infected with the Delta variant reported more symptoms overall, with significantly more symptoms affecting eyes and senses reported. 55% of SARS-CoV-2-positive participants reached a negative N1 Ct value by the day 14 study time point. No risk factors for moderate to severe symptoms were identified in this outpatient cohort. Male sex was associated with a shorter symptom duration. CONCLUSION Symptom manifestation varied among Delta and Omicron variants. Few risk factors were identified for increased symptom severity or duration.
Collapse
Affiliation(s)
- Bhavya Vashi
- The Austere environments Consortium for Enhanced Sepsis Outcomes (ACESO), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Kristen Pettrone
- The Austere environments Consortium for Enhanced Sepsis Outcomes (ACESO), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Claire S. Wilson
- Department of Emergency Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Josh G. Chenoweth
- The Austere environments Consortium for Enhanced Sepsis Outcomes (ACESO), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Joost Brandsma
- The Austere environments Consortium for Enhanced Sepsis Outcomes (ACESO), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Melissa K. Gregory
- The Austere environments Consortium for Enhanced Sepsis Outcomes (ACESO), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Pavol Genzor
- The Austere environments Consortium for Enhanced Sepsis Outcomes (ACESO), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Deborah A. Striegel
- The Austere environments Consortium for Enhanced Sepsis Outcomes (ACESO), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Richard E. Rothman
- Department of Emergency Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Bhakti Hansoti
- Department of Emergency Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Gideon D. Avornu
- Department of Emergency Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Breana McBryde
- Department of Emergency Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Lauren Reynolds Zimmerman
- Department of Emergency Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Christopher W. Woods
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Elizabeth A. Petzold
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Jessica Cowden
- Walter Reed Army Institute of Research-Armed Forces Research Institute of Medical Science (WRAIR-AFRIMS), Bangkok, Thailand
| | - Sorachai Nitayaphan
- Royal Thai Army, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Worapong Nasomsong
- Division of Infectious Disease, Department of Internal Medicine, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| | - Danielle V. Clark
- The Austere environments Consortium for Enhanced Sepsis Outcomes (ACESO), The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| |
Collapse
|
3
|
Erskine F, Spensley K, Prendecki M, Santos E, Anand A, Altmann D, Willicombe M. The Effect of HLA Polymorphism on Immune Response to SARS-CoV-2 Vaccination Within an Infection-Naïve, Vulnerable Population With End-Stage Renal Disease. HLA 2025; 105:e70076. [PMID: 39991976 PMCID: PMC11848999 DOI: 10.1111/tan.70076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/24/2024] [Accepted: 02/04/2025] [Indexed: 02/25/2025]
Abstract
HLA genes exhibit a high degree of polymorphism, contributing to genetic variability known to influence immune responses to infection. Here we investigate associations between HLA polymorphism and serological and T-lymphocyte responses to the BNT162b2 and ChAdOx1 SARS-CoV-2 vaccines within a population receiving maintenance haemodialysis (HD) for End-Stage Renal Disease (ESRD). Our primary objective was to identify HLA alleles associated with diminished serological and T-cellular responsiveness to vaccination. As a secondary objective, the associations between HLA type and COVID-19 disease outcomes were investigated using an independent ESRD cohort (n = 327). This aimed to determine if the alleles associated with poor vaccine response were also linked to unfavourable infection outcomes. In the main study, serum from 225 SARS-CoV-2 infection-naïve patients was HLA-typed using high-resolution Next Generation Sequencing, and serological titres were analysed for the presence of SARS-CoV-2 spike glycoprotein-specific antibodies after two doses of vaccination. A subset of patients (n = 33) was also tested for a T-lymphocyte response. Overall, 89% (n = 200) of patients seroconverted, but only 18% (n = 6) of the cellular response subgroup had a positive T-lymphocyte response. The HLA class II alleles DPB1*104:01, DRB1*04:03 and DRB1*14:04 and HLA class I alleles B*08:01 and B*18:01 were found to significantly correlate with seronegativity, and DQB1*06:01 correlated with serological responsiveness. We were unable to analyse the effect of HLA on disease outcome and T-lymphocyte response due to sample size limitations. Our results suggest pathways for further research and begin to elucidate the relationship between HLA polymorphism and immune responses in the vulnerable ESRD population.
Collapse
Affiliation(s)
- Fiona Erskine
- Imperial College London Department of Surgery and CancerLondonUK
| | - Katrina Spensley
- Imperial College London Department of Surgery and CancerLondonUK
| | - Maria Prendecki
- Imperial College London Department of Surgery and CancerLondonUK
- Imperial College Healthcare NHS TrustLondonUK
| | | | | | - Danny Altmann
- Imperial College London Department of Surgery and CancerLondonUK
| | - Michelle Willicombe
- Imperial College London Department of Surgery and CancerLondonUK
- Imperial College Healthcare NHS TrustLondonUK
| |
Collapse
|
4
|
Feng S, Bibi S, Aley PK, Cappuccini F, Clutterbuck EA, Conlin K, Ebrahimi N, Eordogh A, Faust SN, Felle S, Green J, Gill H, Mujadidi Y, Oladunjoye I, Owino N, Plested E, Robinson H, Stuart A, Voysey M, Pollard AJ, Lambe T. Safety and humoral immunogenicity of the ChAdOx1 nCoV-19 vaccine administered as a fourth dose booster following two doses of ChAdOx1 nCoV-19 and a third dose of BNT162b2 (COV009): A prospective cohort study. J Infect 2025; 90:106423. [PMID: 39826861 DOI: 10.1016/j.jinf.2025.106423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVES Evaluation of the safety and humoral immunogenicity of ChAdOx1 nCoV-19 as a fourth dose booster in individuals who have had two initial doses of the vaccine and a third dose of BNT162b2. METHODS COV009 is a safety follow-up study of volunteers enroled in the pivotal pre-licensure ChAdOx1 nCoV-19. In this sub-study, 149 eligible participants were given a fourth dose of ChAdOx1 nCoV-19. Primary outcomes were reactogenicity, safety, and humoral immunogenicity. Anti-spike IgG and pseudo-neutralising antibody against multiple variants were measured from pre-first dose to 28 days post-second and post-fourth dose (third dose samples were unavailable). RESULTS A fourth dose of ChAdOx1 nCoV-19 had an acceptable safety profile with no vaccine-related serious adverse events. Humoral responses against various SARS CoV-2 variants post-fourth dose were significantly increased compared with the responses after the second dose (7- to 9-fold increase for anti-spike IgG responses across variants, all p<0.05). Those with lower antibody levels prior to the 4th dose had stronger responses to a 4th dose booster. Seropositivity by anti-nucleocapsid, or higher antibody responses pre-fourth dose correlated with lower infection risks six months thereafter (OR: 0.16, 95% CI: 0.05, 0.50). CONCLUSIONS The ChAdOx1 nCoV-19 fourth dose is well tolerated and boosts humoral immunity; this was evident as an increased humoral response across multiple variants of concern. These data support its use as a booster dose against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Shuo Feng
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK; Department of Epidemiology and Biostatistics, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Federica Cappuccini
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Elizabeth A Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Kerry Conlin
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Narges Ebrahimi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Agnes Eordogh
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Saul N Faust
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Trust, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Sally Felle
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Justin Green
- Vaccines and Immune Therapies Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Hardeep Gill
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Yama Mujadidi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Iyiola Oladunjoye
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Nelly Owino
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Emma Plested
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Hannah Robinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Arabella Stuart
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK; Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK; Pandemic Sciences Institute, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
Kirsebom FCM, Hall V, Stowe J. How do large-scale population studies inform vaccine evaluations in England? Clin Exp Immunol 2025; 219:uxaf006. [PMID: 39910973 PMCID: PMC11933692 DOI: 10.1093/cei/uxaf006] [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: 11/05/2024] [Revised: 12/16/2024] [Accepted: 02/04/2025] [Indexed: 02/07/2025] Open
Abstract
Large-scale population studies are important to monitor and evaluate aspects of a vaccination programme including vaccine coverage, real-world effectiveness, and post-licensure vaccine safety. These types of epidemiological studies often come under the remit of public health agencies, such as the UK Health Security Agency (UKHSA) in England, which are required to undertake surveillance of vaccine-preventable diseases, including via seroepidemiological studies and data linkage studies using national-level electronic healthcare data. An individual-level national vaccine register with an accurate denominator can be the key to gaining insights into vaccine coverage, effectiveness, and safety. During the coronavirus disease 2019 pandemic, England's first vaccine register was developed. This enabled timely estimates of real-world vaccine effectiveness in the whole population of England, as well as enabling epidemiological investigations of rare potential risks from vaccines in specific populations. Population-based research studies, including prospective cohort studies, are complementary to surveillance and combined, enable more comprehensive assessments. As there was an unprecedented investment into research studies and infrastructure during the pandemic, the scale of these studies meant they were able to contribute to vaccine programme evaluations in a way that had not been possible for previous vaccine programmes. In this review, we summarise the different large-scale surveillance and research studies that have been used to evaluate and inform vaccine policy from the time of the first data linkage studies undertaken in England in the 1990s to the present-day post-COVID-19 pandemic.
Collapse
Affiliation(s)
- Freja C M Kirsebom
- Immunisation and Vaccine Preventable Diseases Department, UK Health Security Agency, London, UK
| | | | - Julia Stowe
- Immunisation and Vaccine Preventable Diseases Department, UK Health Security Agency, London, UK
| |
Collapse
|
6
|
Mok CKP, Tang YS, Tan CW, Chong KC, Chen C, Sun Y, Yiu K, Ling KC, Chan KK, Hui DS. Comparison of safety and immunogenicity in the elderly after receiving either Comirnaty or Spikevax monovalent XBB1.5 COVID-19 vaccine. J Infect 2025; 90:106374. [PMID: 39657850 DOI: 10.1016/j.jinf.2024.106374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/13/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND The emergence of SARS-CoV-2 variants necessitates ongoing evaluation of vaccine performance. This study evaluates and compares the safety and immunogenicity of the Comirnaty and Spikevax monovalent XBB.1.5 COVID-19 vaccines in an elderly population. METHODS Altogether, 129 elderly individuals were recruited between 2 January and 3 February 2024, and received a booster dose of either Comirnaty (n=59) or Spikevax (n=70) monovalent XBB.1.5 COVID-19 vaccine. Blood samples were collected at before and one month after vaccination. Immunogenicity was assessed by measuring the percentage of IFNγ+CD4+ and IFNγ+CD8+ T cells, and neutralizing antibody titers (NT50) using a surrogate virus neutralization test (sVNT). Adverse reactions were recorded and analyzed. FINDINGS Both vaccines significantly increased the percentage of IFNγ+CD8+ T cells against XBB.1.5 and wild-type (WT) SARS-CoV-2 at one-month post-vaccination. Spikevax induced a significantly higher percentages of IFNγ+CD8+ and CD4+ T cells against XBB.1.5 than Comirnaty (p<0.001). The proportion of participants showing a positive T cell response to XBB1.5 after vaccination was higher in the Spikevax group (64.3% CD8, 71.4% CD4) than in the Comirnaty group (42.4% CD8, 57.6% CD4). Spikevax also elicited higher NT50 levels against XBB1.5, JN.1 and the latest variant KP.2 than Comirnaty (XBB1.5: p<0.01; KP.2: p<0.05). Fever was more common in the Spikevax group (fever: p=0.006). However, all side effects were short-term and resolved on their own. INTERPRETATION Both vaccines induce neutralizing antibody to XBB1.5, JN.1 and KP.2. Specifically, Spikevax induces higher cellular and humoral immune responses than Comirnaty in the elderly, but it is also associated with a higher incidence of fever. These findings can guide public health strategies for vaccinating the elderly population.
Collapse
Affiliation(s)
- Chris Ka Pun Mok
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; SH Ho Research Centre for Emerging Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Yun Sang Tang
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Chee Wah Tan
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore; Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ka Chun Chong
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Centre for Health Systems and Policy Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chunke Chen
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Yuanxin Sun
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Karen Yiu
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Kwun Cheung Ling
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Ken Kp Chan
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - David S Hui
- SH Ho Research Centre for Emerging Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
7
|
Berber E, Ross TM. Factors Predicting COVID-19 Vaccine Effectiveness and Longevity of Humoral Immune Responses. Vaccines (Basel) 2024; 12:1284. [PMID: 39591186 PMCID: PMC11598945 DOI: 10.3390/vaccines12111284] [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: 10/16/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, prompted global efforts to develop vaccines to control the disease. Various vaccines, including mRNA (BNT162b2, mRNA-1273), adenoviral vector (ChAdOx1, Ad26.COV2.S), and inactivated virus platforms (BBIBP-CorV, CoronaVac), elicit high-titer, protective antibodies against the virus, but long-term antibody durability and effectiveness vary. The objective of this study is to elucidate the factors that influence vaccine effectiveness (VE) and the longevity of humoral immune responses to COVID-19 vaccines through a review of the relevant literature, including clinical and real-world studies. Here, we discuss the humoral immune response to different COVID-19 vaccines and identify factors influencing VE and antibody longevity. Despite initial robust immune responses, vaccine-induced immunity wanes over time, particularly with the emergence of variants, such as Delta and Omicron, that exhibit immune escape mechanisms. Additionally, the durability of the humoral immune responses elicited by different vaccine platforms, along with the identification of essential determinants of long-term protection-like pre-existing immunity, booster doses, hybrid immunity, and demographic factors-are critical for protecting against severe COVID-19. Booster vaccinations substantially restore neutralizing antibody levels, especially against immune-evasive variants, while individuals with hybrid immunity have a more durable and potent immune response. Importantly, comorbidities such as diabetes, cardiovascular disease, chronic kidney disease, and cancer significantly reduce the magnitude and longevity of vaccine-induced protection. Immunocompromised individuals, particularly those undergoing chemotherapy and those with hematologic malignancies, have diminished humoral responses and benefit disproportionately from booster vaccinations. Age and sex also influence immune responses, with older adults experiencing accelerated antibody decline and females generally exhibiting stronger humoral responses compared to males. Understanding the variables affecting immune protection is crucial to improving vaccine strategies and predicting VE and protection against COVID-19.
Collapse
Affiliation(s)
- Engin Berber
- Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Ted M. Ross
- Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
- Florida Research and Innovation Center, Cleveland Clinic, Florida, FL 34986, USA
| |
Collapse
|
8
|
Møller M, Friis-Hansen L, Kirkby N, Dilling-Hansen C, Andersson M, Vedsted P, Mølbak K, Koch A. Robust immune response to COVID-19 vaccination in the island population of Greenland. COMMUNICATIONS MEDICINE 2024; 4:173. [PMID: 39242878 PMCID: PMC11379896 DOI: 10.1038/s43856-024-00602-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND In Greenland, the COVID-19 pandemic was characterised by a late onset of community transmission and a low impact on the healthcare system, hypothesised as being partly due to a high uptake of vaccinations. To underpin this description, we aimed to assess the SARS-CoV-2 immune response post-vaccination in a Greenlandic population. METHODS In this observational cohort study, we included 430 adults in Greenland who had received a complete two-dose SARS-CoV-2 vaccination at enrolment. The total plasma SARS-CoV-2 spike glycoprotein Ig antibodies (S-Ab) induced by either the BNT162b2 or mRNA-1273 vaccine, was measured up to 11 months after the second vaccine dose. In addition, total salivary S-Abs were examined in 107 participants, and the T-cell response to the spike glycoprotein was assessed in 78 participants out of the entire study cohort. RESULTS Here we demonstrate that two months after the second vaccine dose, 96% of participants have protective plasma S-Ab levels. By 11 months, 98% have protective levels, with prior SARS-CoV-2 infection particularly enhancing S-Ab levels by 37% (95% CI 25-51%). Among individuals aged 60 years and older, we observe a 21% (95% CI 7-33%) reduction in antibody response. Total salivary S-Ab levels are detectable in all participants and significantly correlate with plasma levels. Moreover, all participants exhibit a robust SARS-CoV-2-specific T-cell response 11 months post-primary vaccination. CONCLUSIONS Our findings show that Greenlanders exhibit a robust and lasting immune response, both humoral and cellular, comparable to other population groups up to at least 11 months after the second vaccine dose. These results corroborate the hypothesis that vaccines contributed to the mild impact of the COVID-19 pandemic in the Greenlandic population.
Collapse
Affiliation(s)
- Mie Møller
- Institue of Health and Nature, University of Greenland, Nuuk, Greenland.
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark.
- Department of Internal Medicine, Queen Ingrid's Hospital, Nuuk, Greenland.
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark.
- Department of Infectious Diseases, Rigshospitalet University Hospital, Copenhagen, Denmark.
| | - Lennart Friis-Hansen
- Department of Clinical Microbiology, Rigshospitalet University Hospital, Copenhagen, Denmark
- Department of Clinical Biochemistry, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Nikolai Kirkby
- Department of Clinical Microbiology, Rigshospitalet University Hospital, Copenhagen, Denmark
| | | | - Mikael Andersson
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Vedsted
- Department of Clinical Medicine, University of Aarhus, Aarhus, Denmark
- Ilulissat Regional Hospital, Ilulissat, Greenland
| | - Kåre Mølbak
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
| | - Anders Koch
- Institue of Health and Nature, University of Greenland, Nuuk, Greenland
- Department of Internal Medicine, Queen Ingrid's Hospital, Nuuk, Greenland
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institut, Copenhagen, Denmark
- Department of Infectious Diseases, Rigshospitalet University Hospital, Copenhagen, Denmark
| |
Collapse
|
9
|
Wiedemann A, Lhomme E, Huchon M, Foucat E, Bérerd-Camara M, Guillaumat L, Yaradouno M, Tambalou J, Rodrigues C, Ribeiro A, Béavogui AH, Lacabaratz C, Thiébaut R, Richert L, Lévy Y. Long-term cellular immunity of vaccines for Zaire Ebola Virus Diseases. Nat Commun 2024; 15:7666. [PMID: 39227399 PMCID: PMC11372064 DOI: 10.1038/s41467-024-51453-z] [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: 04/16/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024] Open
Abstract
Recent Ebola outbreaks underscore the importance of continuous prevention and disease control efforts. Authorized vaccines include Merck's Ervebo (rVSV-ZEBOV) and Johnson & Johnson's two-dose combination (Ad26.ZEBOV/MVA-BN-Filo). Here, in a five-year follow-up of the PREVAC randomized trial (NCT02876328), we report the results of the immunology ancillary study of the trial. The primary endpoint is to evaluate long-term memory T-cell responses induced by three vaccine regimens: Ad26-MVA, rVSV, and rVSV-booster. Polyfunctional EBOV-specific CD4+ T-cell responses increase after Ad26 priming and are further boosted by MVA, whereas minimal responses are observed in the rVSV groups, declining after one year. In-vitro expansion for eight days show sustained EBOV-specific T-cell responses for up to 60 months post-prime vaccination with both Ad26-MVA and rVSV, with no decline. Cytokine production analysis identify shared biomarkers between the Ad26-MVA and rVSV groups. In secondary endpoint, we observed an elevation of pro-inflammatory cytokines at Day 7 in the rVSV group. Finally, we establish a correlation between EBOV-specific T-cell responses and anti-EBOV IgG responses. Our findings can guide booster vaccination recommendations and help identify populations likely to benefit from revaccination.
Collapse
Affiliation(s)
- Aurélie Wiedemann
- Vaccine Research Institute, Université Paris-Est, Créteil, France
- INSERM U955, Institut Mondor de Recherche Biomedicale (IMRB), Team Lévy, Créteil, France
| | - Edouard Lhomme
- Vaccine Research Institute, Université Paris-Est, Créteil, France
- Univ. Bordeaux, INSERM, Institut Bergonié, CHU de Bordeaux, CIC-EC 1401, Euclid/F-CRIN clinical trials platform, Bordeaux, France
- Univ. Bordeaux, Inserm, Population Health Research Center, UMR 1219, INRIA SISTM, Bordeaux, France
| | - Mélanie Huchon
- Vaccine Research Institute, Université Paris-Est, Créteil, France
- Univ. Bordeaux, Inserm, Population Health Research Center, UMR 1219, INRIA SISTM, Bordeaux, France
| | - Emile Foucat
- Vaccine Research Institute, Université Paris-Est, Créteil, France
- INSERM U955, Institut Mondor de Recherche Biomedicale (IMRB), Team Lévy, Créteil, France
| | | | - Lydia Guillaumat
- Vaccine Research Institute, Université Paris-Est, Créteil, France
- INSERM U955, Institut Mondor de Recherche Biomedicale (IMRB), Team Lévy, Créteil, France
| | | | | | - Cécile Rodrigues
- Vaccine Research Institute, Université Paris-Est, Créteil, France
- INSERM U955, Institut Mondor de Recherche Biomedicale (IMRB), Team Lévy, Créteil, France
| | - Alexandre Ribeiro
- Vaccine Research Institute, Université Paris-Est, Créteil, France
- INSERM U955, Institut Mondor de Recherche Biomedicale (IMRB), Team Lévy, Créteil, France
| | - Abdoul Habib Béavogui
- Centre National de Formation et de Recherche en Santé Rurale (CNFRSR), Maferinyah, Guinea
| | - Christine Lacabaratz
- Vaccine Research Institute, Université Paris-Est, Créteil, France
- INSERM U955, Institut Mondor de Recherche Biomedicale (IMRB), Team Lévy, Créteil, France
| | - Rodolphe Thiébaut
- Vaccine Research Institute, Université Paris-Est, Créteil, France
- Univ. Bordeaux, INSERM, Institut Bergonié, CHU de Bordeaux, CIC-EC 1401, Euclid/F-CRIN clinical trials platform, Bordeaux, France
- Univ. Bordeaux, Inserm, Population Health Research Center, UMR 1219, INRIA SISTM, Bordeaux, France
| | - Laura Richert
- Vaccine Research Institute, Université Paris-Est, Créteil, France
- Univ. Bordeaux, INSERM, Institut Bergonié, CHU de Bordeaux, CIC-EC 1401, Euclid/F-CRIN clinical trials platform, Bordeaux, France
- Univ. Bordeaux, Inserm, Population Health Research Center, UMR 1219, INRIA SISTM, Bordeaux, France
| | - Yves Lévy
- Vaccine Research Institute, Université Paris-Est, Créteil, France.
- INSERM U955, Institut Mondor de Recherche Biomedicale (IMRB), Team Lévy, Créteil, France.
- Assistance Publique-Hôpitaux de Paris, Groupe Henri-Mondor Albert-Chenevier, Service Immunologie Clinique, Créteil, France.
| |
Collapse
|
10
|
Townsend L, Marron L, O'Brien K, Walsh C, Domegan L, McGrath J, Kenny C, Fleming C, Bergin C. Investigating incidence of and factors associated with SARS-CoV-2 infection over a nine-month period in a highly-vaccinated healthcare worker cohort. QJM 2024; 117:638-646. [PMID: 38759112 DOI: 10.1093/qjmed/hcae093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/23/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Healthcare workers (HCWs) are at increased risk of SARS-CoV-2 infection. This risk persists despite the implementation of mitigating factors, including vaccination. The ongoing impact of incident SARS-CoV-2 infection and symptomatic COVID-19 disease in vaccinated HCWs is poorly understood. AIM We aimed to describe the epidemiology of incident SARS-CoV-2 infections, as well as investigating the serological, clinical and demographic factors associated with developing infection. DESIGN This was a multi-centre prospective longitudinal study followed a HCW cohort over a nine-month period. METHODS Spike and nucleocapsid SARS-CoV-2 antibodies were measured at enrolment. Vaccination status, demographics and medical history were collated. Incident infection over the study period was recorded. Multivariable regression models investigated factors associated with nucleocapsid antibody status, incident infection and symptomatic infection. RESULTS About 1260 participants took part, of whom n = 1006 were anti-nucleocapsid antibody positive. Negative anti-nucleocapsid antibody was associated with older age and having a known SARS-CoV-2 acquisition risk. There were n = 274 (22%) incident infections, with n = 225 (87%) diagnosed using antigen tests. Incident infections were associated with lower anti-nucleocapsid titres, increased time since previous SARS-CoV-2 infection, and having a known acquisition risk, but were not associated with vaccination status. CONCLUSIONS This study demonstrates a high rate of incident SARS-CoV-2 infection amongst HCWs, despite broad vaccine coverage. There is a shift in diagnostics, from PCR to antigen testing. We identify at-risk groups for incident infection, and these should continue be targeted as part of risk reduction campaigns. Vaccination status and prior infection status alone are not surrogates for protection.
Collapse
Affiliation(s)
- Liam Townsend
- Department of Infectious Diseases, St James's Hospital, Dublin, D08 NHY1, Ireland
| | - Louise Marron
- Health Protection Surveillance Centre, Dublin, D01 A4A3, Ireland
- ECDC Fellowship Programme, Field Epidemiology Path (EPIET), European Centre for Disease Prevention and Control, Stockholm, Gustav III:s Boulevard 40, Stockholm, 169 73 Solna, Sweden
| | - Katie O'Brien
- Health Protection Surveillance Centre, Dublin, D01 A4A3, Ireland
| | - Cathal Walsh
- Health Protection Surveillance Centre, Dublin, D01 A4A3, Ireland
- Department of Biostatistics, Trinity College Dublin, Dublin, College Green, Dublin, D02 PN40, Ireland
| | - Lisa Domegan
- Health Protection Surveillance Centre, Dublin, D01 A4A3, Ireland
| | - Jonathan McGrath
- Department of Infectious Diseases, St James's Hospital, Dublin, D08 NHY1, Ireland
| | - Claire Kenny
- Department of Infectious Diseases, University Hospital Galway, Galway, Newcastle Road, Galway, H91 YR71, Ireland
| | - Catherine Fleming
- Department of Infectious Diseases, University Hospital Galway, Galway, Newcastle Road, Galway, H91 YR71, Ireland
| | - Colm Bergin
- Department of Infectious Diseases, St James's Hospital, Dublin, D08 NHY1, Ireland
- Department of Clinical Medicine, Trinity College Dublin, Dublin, College Green, Dublin, D02 PN40, Ireland
| |
Collapse
|
11
|
Nazareth J, Martin CA, Pan D, Barr IG, Sullivan SG, Peck H, Veli N, Das M, Bryant L, George N, Gohar M, Gray LJ, Teece L, Vail D, Renals V, Karia A, Renals P, Moss P, Tattersall A, Otter AD, Haldar P, Cooper A, Stephenson I, Wiselka MJ, Tang JW, Nellums L, Pareek M. Immunogenicity of concomitant SARS-CoV-2 and influenza vaccination in UK healthcare workers: a prospective longitudinal observational study. THE LANCET REGIONAL HEALTH. EUROPE 2024; 44:101022. [PMID: 39444701 PMCID: PMC11496956 DOI: 10.1016/j.lanepe.2024.101022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 10/25/2024]
Abstract
Background Co-administration of inactivated influenza vaccine (IIV) and SARS-CoV-2 vaccine may impact SARS-CoV-2 vaccine induced humoral immune responses. We aimed to compare IIV and SARS-CoV-2 vaccine induced cellular and humoral immune responses in those receiving concomitant vaccination to those receiving these vaccines separately. Methods We conducted a cohort study between 29th September 2021 and 5th August 2022 in healthcare workers who worked at the local NHS trust and in the surrounding area that were vaccinated with a mRNA SARS-CoV-2 booster and cell-based IIV. We measured haemagglutination inhibition assay (HAI) titres, SARS-CoV-2 anti-spike antibody and SARS-CoV-2 ELISpot count pre-vaccination, 1-month and 6-months post-vaccination and evaluated differences by vaccine strategy. Findings We recruited 420 participants, 234/420 (56%) were vaccinated concomitantly and 186/420 (44%) separately. The 1-month post-vaccination mean fold rise (MFR) in SARS-CoV-2 anti-spike antibodies was lower in those vaccinated concomitantly compared to separately (MFR [95% confidence interval (CI)] 9.7 [8.3, 11.4] vs 12.8 [10.3, 15.9], p = 0.04). After adjustment for age and sex, the adjusted geometric mean ratio (aGMR) remained lower for those vaccinated concomitantly compared to separately (aGMR [95% CI] 0.80 [0.70, 0.92], p = 0.001). At 6-months post-vaccination, we found no statistically significant difference in SARS-CoV-2 anti-spike antibody titres (aGMR [95% CI] 1.09 [0.87, 1.35], p = 0.45). We found no statistically significant correlation between vaccine strategy with SARS-CoV-2 ELISpot count and influenza HAI titres at 1-month and 6-months post-vaccination. Interpretation Our study found that concomitant vaccination with SARS-CoV-2 and IIV has no statistically significant impacts on long-term immunogenicity. Further research is required to understand the underlying mechanisms and assess the clinical significance of reduced anti-spike antibodies in those vaccinated concomitantly. Funding Research and Innovation (UKRI) through the COVID-19 National Core Studies Immunity (NCSi) programme (MC_PC_20060).
Collapse
Affiliation(s)
- Joshua Nazareth
- Department of Respiratory Sciences, University of Leicester, UK
- Department of Infection and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Development Centre for Population Health, University of Leicester, Leicester, UK
| | - Christopher A. Martin
- Department of Respiratory Sciences, University of Leicester, UK
- Department of Infection and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Development Centre for Population Health, University of Leicester, Leicester, UK
| | - Daniel Pan
- Department of Respiratory Sciences, University of Leicester, UK
- Department of Infection and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Development Centre for Population Health, University of Leicester, Leicester, UK
- Li Ka Shing Centre for Health Information and Discovery, Oxford Big Data Institute, University of Oxford, UK
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ian G. Barr
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Sheena G. Sullivan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, University of Melbourne, Melbourne, Australia
- Department of Epidemiology, University of California, Los Angeles, USA
| | - Heidi Peck
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Neyme Veli
- Department of Respiratory Sciences, University of Leicester, UK
| | - Mrinal Das
- Department of Respiratory Sciences, University of Leicester, UK
| | - Luke Bryant
- Department of Respiratory Sciences, University of Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Nisha George
- Department of Respiratory Sciences, University of Leicester, UK
- Department of Infection and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Development Centre for Population Health, University of Leicester, Leicester, UK
| | - Marjan Gohar
- Department of Respiratory Sciences, University of Leicester, UK
- Department of Infection and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Development Centre for Population Health, University of Leicester, Leicester, UK
| | - Laura J. Gray
- Department of Population Health Sciences, University of Leicester, UK
| | - Lucy Teece
- Department of Population Health Sciences, University of Leicester, UK
| | - Denny Vail
- Research Space, University Hospitals of Leicester NHS Trust, UK
| | - Val Renals
- Research Space, University Hospitals of Leicester NHS Trust, UK
| | - Aleesha Karia
- Research Space, University Hospitals of Leicester NHS Trust, UK
| | - Paul Renals
- Research Space, University Hospitals of Leicester NHS Trust, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | | | - Pranab Haldar
- Department of Respiratory Sciences, University of Leicester, UK
- Department of Respiratory Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Andrea Cooper
- Department of Respiratory Sciences, University of Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Iain Stephenson
- Department of Infection and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Martin J. Wiselka
- Department of Respiratory Sciences, University of Leicester, UK
- Department of Infection and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Julian W. Tang
- Department of Respiratory Sciences, University of Leicester, UK
- Department of Clinical Microbiology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Laura Nellums
- Lifespan and Population Health Academic Unit, School of Medicine, University of Nottingham, Nottingham, UK
- College of Population Health, University of New Mexico, NM, USA
| | - Manish Pareek
- Department of Respiratory Sciences, University of Leicester, UK
- Department of Infection and HIV Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
- Development Centre for Population Health, University of Leicester, Leicester, UK
| |
Collapse
|
12
|
Lee SJ, Woo A, Lee JA, Lee Y, Kim HE, Lee JG, Kim SY, Park MS, Jeong SJ. Immunogenicity and Predictive Factors Associated with Poor Response after Severe Acute Respiratory Syndrome Coronavirus 2 Vaccination in Lung Transplant Patients. Vaccines (Basel) 2024; 12:822. [PMID: 39066460 PMCID: PMC11281714 DOI: 10.3390/vaccines12070822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Lung transplant patients are more likely to develop severe coronavirus disease 2019 (COVID-19) compared with the general population and should be vaccinated against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, previous studies have reported reduced vaccination immunogenicity in lung transplantation patients. We aimed to investigate the serological response and associated factors after SARS-CoV-2 vaccination in this population. Lung transplant patients without a history of contracting coronavirus disease who had received a second or higher dose of SARS-CoV-2 vaccination were enrolled. The anti-SARS-Cov-2 spike and neutralizing antibody levels were measured in blood samples. Firth's logistic regression analysis was performed to assess the factors associated with non-response after vaccination. Forty-six lung transplant patients were enrolled, of which sixteen (34.8%) showed a serological response to vaccination. All patients who received anti-SARS-CoV-2 vaccination before transplantation (n = 5) exhibited a serological response. No significant difference was observed in anti-SARS-CoV-2 S antibody or neutralization titers based on the number and timing of vaccination. Firth's logistic regression showed an association between lower hemoglobin levels (odds ratio, 0.59; confidence interval, 0.35-0.92; p = 0.017) and non-response to SARS-CoV-2 vaccination. Lung transplant patients showed poor serologic responses after SARS-CoV-2 vaccination in this pilot study; anemia may be associated with this poor response.
Collapse
Affiliation(s)
- Se Ju Lee
- Division of Infectious Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.J.L.); (J.A.L.); (Y.L.)
- Division of Infectious Diseases, Department of Internal Medicine, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Ala Woo
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (A.W.); (S.Y.K.); (M.S.P.)
| | - Jung Ah Lee
- Division of Infectious Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.J.L.); (J.A.L.); (Y.L.)
| | - Yongseop Lee
- Division of Infectious Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.J.L.); (J.A.L.); (Y.L.)
| | - Ha Eun Kim
- Department of Thoracic and Cardiovascular Surgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (H.E.K.); (J.G.L.)
| | - Jin Gu Lee
- Department of Thoracic and Cardiovascular Surgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (H.E.K.); (J.G.L.)
| | - Song Yee Kim
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (A.W.); (S.Y.K.); (M.S.P.)
| | - Moo Suk Park
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (A.W.); (S.Y.K.); (M.S.P.)
| | - Su Jin Jeong
- Division of Infectious Diseases, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.J.L.); (J.A.L.); (Y.L.)
| |
Collapse
|
13
|
Hirai K, Shimotashiro M, Okumura T, Ookawara S, Morishita Y. Anti-SARS-CoV-2 spike antibody response to the third dose of BNT162b2 mRNA COVID-19 vaccine and associated factors in Japanese hemodialysis patients. Kidney Res Clin Pract 2024; 43:326-336. [PMID: 38389151 PMCID: PMC11181050 DOI: 10.23876/j.krcp.23.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 09/02/2023] [Accepted: 09/19/2023] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND We assessed the anti-SARS-CoV-2 spike antibody response to the third dose of BNT162b2 mRNA COVID-19 vaccine in Japanese hemodialysis patients and determined factors associated with the anti-SARS-CoV-2 spike antibody titer after the third dose of COVID-19 vaccine. METHODS Overall, 64 patients were enrolled in this single-center, prospective, longitudinal study. Anti-SARS-CoV-2 spike antibody titers were compared between hemodialysis patients and 18 healthcare workers. Multiple linear regression analysis was used to identify factors associated with the anti-SARS-CoV-2 spike antibody titer after the third vaccination. RESULTS There was no significant difference in anti-SARS-CoV-2 spike antibody titer 4 weeks after the third vaccination between hemodialysis patients and healthcare workers (18,500 [interquartile range, 11,000-34,500] vs. 11,500 [interquartile range, 7,918- 19,500], all values in AU/mL; p = 0.17). Uric acid (standard coefficient [β] = -0.203, p = 0.02), transferrin saturation (β = -0.269, p = 0.003), and log-anti-SARS-CoV-2 spike antibody titer 1 week before the third vaccination (β = 0.440, p < 0.001) correlated with the log-anti-SARS-CoV-2 spike antibody titer 4 weeks after the third vaccination. In contrast, only the log-anti-SARS-CoV-2 spike antibody titer 1 week before the third vaccination (β = 0.410, p < 0.001) correlated with the log- anti-SARS-CoV-2 spike antibody titer 12 weeks after the third vaccination. CONCLUSION The anti-SARS-CoV-2 spike antibody titer after the third dose of COVID-19 vaccine was comparable between hemodialysis patients and healthcare workers. Uric acid concentration, transferrin saturation, and anti-SARS-CoV-2 spike antibody titer before the third dose were associated with the anti-SARS-CoV-2 spike antibody titer after the third dose in Japanese hemodialysis patients.
Collapse
Affiliation(s)
- Keiji Hirai
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | | | | | - Susumu Ookawara
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Yoshiyuki Morishita
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| |
Collapse
|
14
|
Santos-Rebouças CB, Ferreira CDS, Nogueira JDS, Brustolini OJ, de Almeida LGP, Gerber AL, Guimarães APDC, Piergiorge RM, Struchiner CJ, Porto LC, de Vasconcelos ATR. Immune response stability to the SARS-CoV-2 mRNA vaccine booster is influenced by differential splicing of HLA genes. Sci Rep 2024; 14:8982. [PMID: 38637586 PMCID: PMC11026523 DOI: 10.1038/s41598-024-59259-1] [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: 09/12/2023] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
Many molecular mechanisms that lead to the host antibody response to COVID-19 vaccines remain largely unknown. In this study, we used serum antibody detection combined with whole blood RNA-based transcriptome analysis to investigate variability in vaccine response in healthy recipients of a booster (third) dose schedule of the mRNA BNT162b2 vaccine against COVID-19. The cohort was divided into two groups: (1) low-stable individuals, with antibody concentration anti-SARS-CoV IgG S1 below 0.4 percentile at 180 days after boosting vaccination; and (2) high-stable individuals, with antibody values greater than 0.6 percentile of the range in the same period (median 9525 [185-80,000] AU/mL). Differential gene expression, expressed single nucleotide variants and insertions/deletions, differential splicing events, and allelic imbalance were explored to broaden our understanding of the immune response sustenance. Our analysis revealed a differential expression of genes with immunological functions in individuals with low antibody titers, compared to those with higher antibody titers, underscoring the fundamental importance of the innate immune response for boosting immunity. Our findings also provide new insights into the determinants of the immune response variability to the SARS-CoV-2 mRNA vaccine booster, highlighting the significance of differential splicing regulatory mechanisms, mainly concerning HLA alleles, in delineating vaccine immunogenicity.
Collapse
Affiliation(s)
- Cíntia Barros Santos-Rebouças
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Cristina Dos Santos Ferreira
- Bioinformatics Laboratory-LABINFO, National Laboratory of Scientific Computation LNCC/MCTIC, Getúlio Vargas, Av., 333, Quitandinha, Petrópolis, Rio de Janeiro, 25651‑075, Brazil
| | - Jeane de Souza Nogueira
- Histocompatibility and Cryopreservation Laboratory, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Otávio José Brustolini
- Bioinformatics Laboratory-LABINFO, National Laboratory of Scientific Computation LNCC/MCTIC, Getúlio Vargas, Av., 333, Quitandinha, Petrópolis, Rio de Janeiro, 25651‑075, Brazil
| | - Luiz Gonzaga Paula de Almeida
- Bioinformatics Laboratory-LABINFO, National Laboratory of Scientific Computation LNCC/MCTIC, Getúlio Vargas, Av., 333, Quitandinha, Petrópolis, Rio de Janeiro, 25651‑075, Brazil
| | - Alexandra Lehmkuhl Gerber
- Bioinformatics Laboratory-LABINFO, National Laboratory of Scientific Computation LNCC/MCTIC, Getúlio Vargas, Av., 333, Quitandinha, Petrópolis, Rio de Janeiro, 25651‑075, Brazil
| | - Ana Paula de Campos Guimarães
- Bioinformatics Laboratory-LABINFO, National Laboratory of Scientific Computation LNCC/MCTIC, Getúlio Vargas, Av., 333, Quitandinha, Petrópolis, Rio de Janeiro, 25651‑075, Brazil
| | - Rafael Mina Piergiorge
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Cláudio José Struchiner
- School of Applied Mathematics, Getúlio Vargas Foundation, Rio de Janeiro, Brazil
- Social Medicine Institute Hesio Cordeiro, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Luís Cristóvão Porto
- Histocompatibility and Cryopreservation Laboratory, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Ana Tereza Ribeiro de Vasconcelos
- Bioinformatics Laboratory-LABINFO, National Laboratory of Scientific Computation LNCC/MCTIC, Getúlio Vargas, Av., 333, Quitandinha, Petrópolis, Rio de Janeiro, 25651‑075, Brazil.
| |
Collapse
|
15
|
Kim JS, Sun Y, Balte P, Cushman M, Boyle R, Tracy RP, Styer LM, Bell TD, Anderson MR, Allen NB, Schreiner PJ, Bowler RP, Schwartz DA, Lee JS, Xanthakis V, Doyle MF, Regan EA, Make BJ, Kanaya AM, Wenzel SE, Coresh J, Isasi CR, Raffield LM, Elkind MSV, Howard VJ, Ortega VE, Woodruff P, Cole SA, Henderson JM, Mantis NJ, Parker MM, Demmer RT, Oelsner EC. Demographic and Clinical Factors Associated With SARS-CoV-2 Spike 1 Antibody Response Among Vaccinated US Adults: the C4R Study. Nat Commun 2024; 15:1492. [PMID: 38374032 PMCID: PMC10876680 DOI: 10.1038/s41467-024-45468-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 01/24/2024] [Indexed: 02/21/2024] Open
Abstract
This study investigates correlates of anti-S1 antibody response following COVID-19 vaccination in a U.S. population-based meta-cohort of adults participating in longstanding NIH-funded cohort studies. Anti-S1 antibodies were measured from dried blood spots collected between February 2021-August 2022 using Luminex-based microsphere immunoassays. Of 6245 participants, mean age was 73 years (range, 21-100), 58% were female, and 76% were non-Hispanic White. Nearly 52% of participants received the BNT162b2 vaccine and 48% received the mRNA-1273 vaccine. Lower anti-S1 antibody levels are associated with age of 65 years or older, male sex, higher body mass index, smoking, diabetes, COPD and receipt of BNT16b2 vaccine (vs mRNA-1273). Participants with a prior infection, particularly those with a history of hospitalized illness, have higher anti-S1 antibody levels. These results suggest that adults with certain socio-demographic and clinical characteristics may have less robust antibody responses to COVID-19 vaccination and could be prioritized for more frequent re-vaccination.
Collapse
Affiliation(s)
- John S Kim
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Yifei Sun
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Pallavi Balte
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Mary Cushman
- Department of Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Rebekah Boyle
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Linda M Styer
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Taison D Bell
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | | | - Norrina B Allen
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pamela J Schreiner
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Russell P Bowler
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - David A Schwartz
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Joyce S Lee
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Vanessa Xanthakis
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Margaret F Doyle
- Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | | | - Barry J Make
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - Alka M Kanaya
- Division of General Internal Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sally E Wenzel
- Department of Medicine, Department of Immunology, and Department of Environmental Medicine and Occupational Health, University of Pittsburgh School of Medicine, School of Public Health, Pittsburgh, PA, USA
| | - Josef Coresh
- Department of Population Health, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
- Department of Medicine, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Carmen R Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Mitchell S V Elkind
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Virginia J Howard
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor E Ortega
- Division of Respiratory Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Prescott Woodruff
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Shelley A Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Joel M Henderson
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, USA
| | - Monica M Parker
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA.
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA.
- Division of Epidemiology, Department of Quantitative Health Sciences, College of Medicine and Science, Mayo Clinic, Rochester, MN, USA.
| | - Elizabeth C Oelsner
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
16
|
Ankunda V, Katende JS, Oluka GK, Sembera J, Baine C, Odoch G, Ejou P, Kato L, Kaleebu P, Serwanga J. The subdued post-boost spike-directed secondary IgG antibody response in Ugandan recipients of the Pfizer-BioNTech BNT162b2 vaccine has implications for local vaccination policies. Front Immunol 2024; 15:1325387. [PMID: 38469296 PMCID: PMC10926532 DOI: 10.3389/fimmu.2024.1325387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction This study aimed to delineate longitudinal antibody responses to the Pfizer-BioNTech BNT162b2 COVID-19 vaccine within the Ugandan subset of the Sub-Saharan African (SSA) demographic, filling a significant gap in global datasets. Methods We enrolled 48 participants and collected 320 specimens over 12 months after the primary vaccination dose. A validated enzyme-linked immunosorbent assay (ELISA) was used to quantify SARS-CoV-2-specific IgG, IgM, and IgA antibody concentrations (ng/ml) and optical densities (ODs). Statistical analyses included box plots, diverging bar graphs, and the Wilcoxon test with Bonferroni correction. Results We noted a robust S-IgG response within 14 days of the primary vaccine dose, which was consistent with global data. There was no significant surge in S-IgG levels after the booster dose, contrasting trends in other global populations. The S-IgM response was transient and predominantly below established thresholds for this population, which reflects its typical early emergence and rapid decline. S-IgA levels rose after the initial dose then decreased after six months, aligning with the temporal patterns of mucosal immunity. Eleven breakthrough infections were noted, and all were asymptomatic, regardless of the participants' initial S-IgG serostatus, which suggests a protective effect from vaccination. Discussion The Pfizer-BioNTech BNT162b2 COVID-19 vaccine elicited strong S-IgG responses in the SSA demographic. The antibody dynamics distinctly differed from global data highlighting the significance of region-specific research and the necessity for customised vaccination strategies.
Collapse
Affiliation(s)
- Violet Ankunda
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Joseph Ssebwana Katende
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Gerald Kevin Oluka
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Jackson Sembera
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Claire Baine
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Geoffrey Odoch
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Peter Ejou
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Laban Kato
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Pontiano Kaleebu
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Jennifer Serwanga
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| |
Collapse
|
17
|
Asamoah-Boaheng M, Grunau B, Karim ME, Kayda I, Yap J, Bessai K, Goldfarb DM. Investigating the Antibody Imprinting Hypothesis among Canadian Paramedics after SARS-CoV-2 Omicron Variant Circulation. Immunohorizons 2024; 8:193-197. [PMID: 38386593 PMCID: PMC10916361 DOI: 10.4049/immunohorizons.2400010] [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: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Recent research has highlighted the Omicron variant's capacity to evade immune protection conferred by wild-type (WT) mRNA vaccines. Despite this observation, the potential involvement of antigenic sin phenomena remains unclear. Our hypothesis posited that a greater number of prior WT vaccine doses might lead to reduced anti-Omicron neutralization Abs following Omicron infection. To investigate this, we analyzed blood samples from human participants in the COVID-19 Occupational Risk, Seroprevalence, and Immunity among Paramedics (CORSIP) study who had received at least one WT mRNA vaccine before contracting Omicron. The exposure variable was the number of WT mRNA vaccines administered, and the outcome was the angiotensin-converting enzyme 2 (ACE-2) percent inhibition specific to the BA.4/BA.5 Omicron Ag. Contrary to expectations, our findings revealed that more WT-based vaccines were associated with an enhanced Omicron-specific immune response.
Collapse
Affiliation(s)
- Michael Asamoah-Boaheng
- Department of Emergency Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Advancing Health Outcomes, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Resuscitation Research Collaborative, Vancouver, British Columbia, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian Grunau
- Department of Emergency Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Advancing Health Outcomes, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Resuscitation Research Collaborative, Vancouver, British Columbia, Canada
| | - Mohammad Ehsanul Karim
- Centre for Advancing Health Outcomes, University of British Columbia, Vancouver, British Columbia, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Iryna Kayda
- Experimental Medicine Graduate Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Justin Yap
- British Columbia Resuscitation Research Collaborative, Vancouver, British Columbia, Canada
- Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katherine Bessai
- British Columbia Emergency Health Services, British Columbia, Canada
| | - David M. Goldfarb
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
18
|
Datwani S, Kalikawe R, Mwimanzi F, Speckmaier S, Liang R, Sang Y, Waterworth R, Yaseen F, Lapointe HR, Barad E, DeMarco ML, Holmes DT, Simons J, Montaner JS, Romney MG, Brumme ZL, Brockman MA. Dynamics of T-cell Responses Following COVID-19 mRNA Vaccination and Breakthrough Infection in Older Adults. Pathog Immun 2023; 8:117-135. [PMID: 38035132 PMCID: PMC10686373 DOI: 10.20411/pai.v8i1.613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/18/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction While older adults generally mount weaker antibody responses to a primary COVID-19 vaccine series, T-cell responses remain less well characterized in this population. We compared SARS-CoV-2 spike-specific T-cell responses after 2- and 3-dose COVID-19 mRNA vaccination and subsequent breakthrough infection in older and younger adults. Methods We quantified CD4+ and CD8+ T-cells reactive to overlapping peptides spanning the ancestral SARS-CoV-2 spike protein in 40 older adults (median age 79) and 50 younger health care workers (median age 39), all COVID-19 naive, using an activation-induced marker assay. T-cell responses were further assessed in 24 participants, including 8 older adults, who subsequently experienced their first SARS-CoV-2 breakthrough infection. Results A third COVID-19 mRNA vaccine dose significantly boosted spike-specific CD4+ and CD8+ T-cell frequencies to above 2-dose levels in older and younger adults. T-cell frequencies did not significantly differ between older and younger adults after either dose. Multivariable analyses adjusting for sociodemographic, health, and vaccine-related variables confirmed that older age was not associated with impaired cellular responses. Instead, the strongest predictors of CD4+ and CD8+ T-cell frequencies post-third-dose were their corresponding post-second-dose frequencies. Breakthrough infection significantly increased both CD4+ and CD8+ T-cell frequencies, to comparable levels in older and younger adults. Exploratory analyses revealed an association between HLA-A*02:03 and higher post-vaccination CD8+ T-cell frequencies, which may be attributable to numerous strong-binding HLA-A*02:03-specific CD8+ T-cell epitopes in the spike protein. Conclusion Older adults mount robust T-cell responses to 2- and 3-dose COVID-19 mRNA vaccination, which are further boosted following breakthrough infection.
Collapse
Affiliation(s)
- Sneha Datwani
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Rebecca Kalikawe
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Francis Mwimanzi
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Sarah Speckmaier
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Richard Liang
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Yurou Sang
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Rachel Waterworth
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Fatima Yaseen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Hope R. Lapointe
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Evan Barad
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Mari L. DeMarco
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Daniel T. Holmes
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Janet Simons
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Julio S.G. Montaner
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Marc G. Romney
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Zabrina L. Brumme
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Mark A. Brockman
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
19
|
Costiniuk CT, Singer J, Lee T, Galipeau Y, McCluskie PS, Arnold C, Langlois MA, Needham J, Jenabian MA, Burchell AN, Samji H, Chambers C, Walmsley S, Ostrowski M, Kovacs C, Tan DH, Harris M, Hull M, Brumme ZL, Lapointe HR, Brockman MA, Margolese S, Mandarino E, Samarani S, Vulesevic B, Lebouché B, Angel JB, Routy JP, Cooper CL, Anis AH. Antibody neutralization capacity after coronavirus disease 2019 vaccination in people with HIV in Canada. AIDS 2023; 37:F25-F35. [PMID: 37534695 PMCID: PMC10481923 DOI: 10.1097/qad.0000000000003680] [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: 07/07/2023] [Accepted: 07/15/2023] [Indexed: 08/04/2023]
Abstract
OBJECTIVES Many vaccines require higher/additional doses or adjuvants to provide adequate protection for people with HIV (PWH). Here, we compare coronavirus disease 2019 (COVID-19) vaccine-induced antibody neutralization capacity in PWH vs. HIV-negative individuals following two vaccine doses. DESIGN In Canadian prospective observational cohorts, including a multicentre study of PWH receiving at least two COVID-19 vaccinations (mRNA or ChAdOx1-S), and a parallel study of HIV-negative controls (Stop the Spread Ottawa Cohort), we measured vaccine-induced neutralization capacity 3 months post dose 2 (±1 month). METHODS COVID-19 neutralization efficiency was measured by calculating the half maximal inhibitory dilution (ID50) using a high-throughput protein-based neutralization assay for Ancestral (Wuhan), Delta and Omicron (BA.1) spike variants. Univariable and multivariable quantile regression were used to compare COVID-19-specific antibody neutralization capacity by HIV status. RESULTS Neutralization assays were performed on 256 PWH and 256 controls based on specimen availability at the timepoint of interest, having received two vaccines and known date of vaccination. There was a significant interaction between HIV status and previous COVID-19 infection status in median ID50. There were no differences in median ID50 for HIV+ vs. HIV-negative persons without past COVID-19 infection. For participants with past COVID-19 infection, median ICD50 was significantly higher in controls than in PWH for ancestral SARS-CoV-2 and Omicron variants, with a trend for the Delta variant in the same direction. CONCLUSION Vaccine-induced SARS-CoV-2 neutralization capacity was similar between PWH vs. HIV-negative persons without past COVID-19 infection, demonstrating favourable humoral-mediated immunogenicity. Both HIV+ and HIV-negative persons demonstrated hybrid immunity. TRIAL REGISTRATION clinicaltrials.gov NCT04894448.
Collapse
Affiliation(s)
- Cecilia T. Costiniuk
- Division of Infectious Diseases/Chronic Viral Illness Service, McGill University Health Centre, Royal Victoria Hospital
- Infectious Diseases and Immunity in Global Health Research Program, Research Institute of McGill University Health Centre
- Department of Experimental Medicine, McGill University, Montreal, Québec
| | - Joel Singer
- School of Population and Public Health, University of British Columbia
- CIHR Canadian HIV Trials Network (CTN)
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, British Columbia
| | - Terry Lee
- CIHR Canadian HIV Trials Network (CTN)
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, British Columbia
| | - Yannick Galipeau
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario
| | - Pauline S. McCluskie
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario
| | - Corey Arnold
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario
| | - Judy Needham
- CIHR Canadian HIV Trials Network (CTN)
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, British Columbia
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, Québec
| | - Ann N. Burchell
- Department of Family and Community Medicine, St Michael's Hospital, Unity Health Toronto
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario
| | - Hasina Samji
- Faculty of Health Sciences, Simon Fraser University, Burnaby
- British Columbia Centre for Disease Control, Vancouver, British Columbia
| | - Catharine Chambers
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario
- MAP Centre for Urban Health Solutions, St Michael's Hospital
| | - Sharon Walmsley
- Division of Infectious Diseases, Department of Medicine, University of Toronto
| | - Mario Ostrowski
- Clinical Sciences Division and Department of Immunology, University of Toronto, Li Ka Shing Knowledge Institute, St. Michael's Hospital
| | | | - Darrell H.S. Tan
- MAP Centre for Urban Health Solutions, St Michael's Hospital
- Division of Infectious Diseases, Department of Medicine, University of Toronto
- Institute of Public Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario
| | - Marianne Harris
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver
| | - Mark Hull
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver
| | - Zabrina L. Brumme
- Faculty of Health Sciences, Simon Fraser University, Burnaby
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver
| | | | - Mark A. Brockman
- Faculty of Health Sciences, Simon Fraser University, Burnaby
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver
- Department of Molecular Biology and Biochemistry, Faculty of Science, Simon Fraser University, Burnaby, British Columbia
| | | | | | - Suzanne Samarani
- Division of Infectious Diseases/Chronic Viral Illness Service, McGill University Health Centre, Royal Victoria Hospital
| | - Branka Vulesevic
- CIHR Canadian HIV Trials Network (CTN)
- Division of Infectious Diseases, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, Ontario
| | - Bertrand Lebouché
- Division of Infectious Diseases/Chronic Viral Illness Service, McGill University Health Centre, Royal Victoria Hospital
- Infectious Diseases and Immunity in Global Health Research Program, Research Institute of McGill University Health Centre
- Department of Family Medicine, Faculty of Medicine and Health Sciences, McGill University
- Canadian Institutes of Health Research Strategy for Patient-Oriented Research Mentorship Chair in Innovative Clinical Trials
| | - Jonathan B. Angel
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario
- Division of Infectious Diseases, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, Ontario
| | - Jean-Pierre Routy
- Division of Infectious Diseases/Chronic Viral Illness Service, McGill University Health Centre, Royal Victoria Hospital
- Infectious Diseases and Immunity in Global Health Research Program, Research Institute of McGill University Health Centre
- Division of Hematology, Department of Medicine, McGill University Health Centre, Montreal, Québec, Canada
| | - Curtis L. Cooper
- Division of Infectious Diseases, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, Ontario
| | - Aslam H. Anis
- School of Population and Public Health, University of British Columbia
- CIHR Canadian HIV Trials Network (CTN)
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, British Columbia
| |
Collapse
|