1
|
Lürken K, Meinecke A, Manthey LA, Cossmann A, Stankov MV, Klawonn F, Zychlinsky Scharff A, Steffens S, Dopfer-Jablonka A, Müller F, Behrens GMN, Happle C. Impaired Hepatitis B and COVID-19 vaccination responses show strong concordance in hemodialysis patients with end stage renal disease. Eur J Med Res 2025; 30:34. [PMID: 39819737 PMCID: PMC11736940 DOI: 10.1186/s40001-025-02274-3] [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: 11/12/2024] [Accepted: 01/04/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Patients with end stage renal disease (ESRD) undergoing hemodialysis are at increased risk for infection and impaired vaccination responses. We analyzed overlap and influencing factors of vaccination responses against severe acute respiratory syndrome corona virus disease 2 (SARS-CoV-2) and Hepatitis B virus (HBV). METHODS SARS-CoV-2 and HBV vaccination response was assessed in a cohort of German ESRD hemodialysis patients. Anti-HBs- and SARS-CoV-2 anti-S-IgG were analyzed by ELISA. Demographic and clinical data were extracted from clinical files. RESULTS Sixty-four patients with complete information on HBV and SARS-CoV-2 vaccination responses were included. More than one-third (35.4%) of non-responders upon HBV vaccination were identified. Unresponsiveness after HBV and poor response after SARS-CoV-2 vaccination showed strong overlap, and overall, 70.3% of patients were classified into concordant HBV/SARS vaccination response groups. HBV vaccination non-responsiveness, but not poor SARS-CoV-2 post-vaccination immunity was associated with obesity, while poor SARS-CoV-2 vaccination responses were associated increased age. CONCLUSION Our findings confirm previous reports on impaired vaccination response in hemodialysis patients and show that post-vaccination humoral responses against SARS-CoV-2 and HBV display strong overlap in this vulnerable patient group. These results may help to adapt vaccination strategies in this highly vulnerable population. TRIAL REGISTRATION German Clinical Trial Registry, DRKS00021152.
Collapse
Affiliation(s)
- Karsten Lürken
- Dialysis Centre Eickenhof, Langenhagen, Germany
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Anna Meinecke
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Luis A Manthey
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Anne Cossmann
- Dialysis Centre Eickenhof, Langenhagen, Germany
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Metodi V Stankov
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Frank Klawonn
- Institute of Information Engineering, Ostfalia University of Applied Sciences, Wolfenbüttel, Germany
- Biostatistics Research Group, Helmholtz-Center for Infection Research, Braunschweig, Germany
| | - Anna Zychlinsky Scharff
- Department of Pediatric Pneumology, Allergology, and Neonatology, Hannover Medical School, Hannover, Germany
- Department of Pediatric Oncology and Hematology, Hannover Medical School, Hannover, Germany
| | - Sandra Steffens
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Alexandra Dopfer-Jablonka
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Frank Müller
- Department of General Practice, University Medical Center Göttingen, Göttingen, Germany
- Department of Family Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Georg M N Behrens
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
- CiiM - Centre for Individualized Infection Medicine, Hannover, Germany
| | - Christine Happle
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany.
- Department of Pediatric Pneumology, Allergology, and Neonatology, Hannover Medical School, Hannover, Germany.
- Biomedical Research in End Stage and Obstructive Lung Disease/BREATH Hannover, German Centre for Lung Research (DZL), Hannover, Germany.
| |
Collapse
|
2
|
Paredes M, Cruzado P, Bellver C, Ruiz-Albert Y, Cuevas E, Suárez-Cuartin G, Córdoba-Izquierdo A, Sarasate M, Santos S, Gasa M. Learnings From SARS-CoV-2 Patients in an Intermediate Respiratory Care Unit Regardless the Stated Therapeutic Ceiling. OPEN RESPIRATORY ARCHIVES 2025; 7:100373. [PMID: 39717142 PMCID: PMC11663952 DOI: 10.1016/j.opresp.2024.100373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Affiliation(s)
- Marc Paredes
- Respiratory Department, Bellvitge Biomedical Research Institute (IDIBELL), Bellvitge University Hospital, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Pau Cruzado
- Respiratory Department, Bellvitge Biomedical Research Institute (IDIBELL), Bellvitge University Hospital, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Bellver
- Respiratory Department, Bellvitge Biomedical Research Institute (IDIBELL), Bellvitge University Hospital, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Yolanda Ruiz-Albert
- Respiratory Department, Bellvitge Biomedical Research Institute (IDIBELL), Bellvitge University Hospital, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Ester Cuevas
- Respiratory Department, Bellvitge Biomedical Research Institute (IDIBELL), Bellvitge University Hospital, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Guillermo Suárez-Cuartin
- Respiratory Department, Bellvitge Biomedical Research Institute (IDIBELL), Bellvitge University Hospital, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
- Department of Medicine, Campus Bellvitge, Universitat de Barcelona, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Ana Córdoba-Izquierdo
- Respiratory Department, Bellvitge Biomedical Research Institute (IDIBELL), Bellvitge University Hospital, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Mikel Sarasate
- Respiratory Department, Bellvitge Biomedical Research Institute (IDIBELL), Bellvitge University Hospital, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Salud Santos
- Respiratory Department, Bellvitge Biomedical Research Institute (IDIBELL), Bellvitge University Hospital, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
- Department of Medicine, Campus Bellvitge, Universitat de Barcelona, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| | - Mercè Gasa
- Respiratory Department, Bellvitge Biomedical Research Institute (IDIBELL), Bellvitge University Hospital, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
- Department of Medicine, Campus Bellvitge, Universitat de Barcelona, 08907 L’Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
3
|
Cai G, Liu S, Lu Y, Takaki Y, Matsumoto F, Yoshikawa A, Taguri T, Xie J, Arima K, Mizukami S, Wu J, Yamamoto T, Hasegawa M, Tien Huy N, Saito M, Takeuchi S, Morita K, Aoyagi K, He F. Impact of COVID-19 vaccination status on hospitalization and disease severity: A descriptive study in Nagasaki Prefecture, Japan. Hum Vaccin Immunother 2024; 20:2322795. [PMID: 38517220 PMCID: PMC10962621 DOI: 10.1080/21645515.2024.2322795] [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: 10/25/2023] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) was extraordinarily harmful, with high rates of infection and hospitalization. This study aimed to evaluate the impact of COVID-19 vaccination status and other factors on hospitalization and disease severity, using data from Nagasaki Prefecture, Japan. Confirmed cases of COVID-19 infection with vaccination status were included and the differences in characteristics between different vaccination statuses, hospitalization or not, and patients with varying levels of disease severity were analyzed. Furthermore, logistic regression was used to calculate odds ratio (ORs) and 95% confidence intervals (CI) to evaluate the association of various factors with hospitalization and disease severity. From March 14, 2020 to August 31, 2022, 23,139 patients were unvaccinated 13,668 vaccinated the primary program with one or two doses, and 4,575 completed the booster. Vaccination reduced the risk of hospitalization with an odd ratio of 0.759 (95% CI: 0.654-0.881) and the protective effect of completed booster vaccination was more pronounced (OR: 0.261, 95% CI: 0.207-0.328). Similarly, vaccination significantly reduced the risk of disease severity (vaccinated primary program: OR: 0.191, 95% CI: 0.160-0.228; completed booster vaccination: OR: 0.129, 95% CI: 0.099-0.169). Overall, unvaccinated, male, elderly, immunocompromised, obese, and patients with other severe illness factors were all risk factors for COVID-19-related hospitalization and disease severity. Vaccination was associated with a decreased risk of hospitalization and disease severity, and highlighted the benefits of completing booster.
Collapse
Affiliation(s)
- Guoxi Cai
- Public Health and Hygiene Research Department, Nagasaki Prefectural Institute of Environment and Public Health, Nagasaki, Japan
- Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of International Health and Medical Anthropology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Shiwen Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, 1 Xuefu North Road, Fuzhou, Fujian Province, China
| | - Yixiao Lu
- Department of Systems Biology and Health Statistics, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yumika Takaki
- Public Health and Hygiene Research Department, Nagasaki Prefectural Institute of Environment and Public Health, Nagasaki, Japan
| | - Fumiaki Matsumoto
- Public Health and Hygiene Research Department, Nagasaki Prefectural Institute of Environment and Public Health, Nagasaki, Japan
| | - Akira Yoshikawa
- Public Health and Hygiene Research Department, Nagasaki Prefectural Institute of Environment and Public Health, Nagasaki, Japan
| | - Toshitsugu Taguri
- Public Health and Hygiene Research Department, Nagasaki Prefectural Institute of Environment and Public Health, Nagasaki, Japan
| | - Jianfen Xie
- Fujian Provincial Center for Disease Control and Prevention, Fuzhou, Fujian Province, China
| | - Kazuhiko Arima
- Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Satoshi Mizukami
- Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Jiwen Wu
- Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Taro Yamamoto
- Department of International Health and Medical Anthropology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Maiko Hasegawa
- Infectious Disease Control Office, Health & Welfare Department, Nagasaki Prefectural Government, Nagasaki, Japan
| | - Nguyen Tien Huy
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- School of Medicine and Pharmacy, Duy Tan University, Da Nang, Vietnam
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Masaya Saito
- Department of Nutrition Science, Faculty of Nursing and Nutrition, University of Nagasaki, Nagasaki, Japan
| | - Shouhei Takeuchi
- Department of Nutrition Science, Faculty of Nursing and Nutrition, University of Nagasaki, Nagasaki, Japan
| | - Kouichi Morita
- Department of Virology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- Dejima Infectious Disease Research Alliance, Nagasaki University, Nagasaki, Japan
| | - Kiyoshi Aoyagi
- Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Fei He
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, 1 Xuefu North Road, Fuzhou, Fujian Province, China
- Fujian Provincial Key Laboratory of Tumor Microbiology, Fujian Medical University, Fujian Province, China
- Fujian Digital Tumor Data Research Center, Fujian Province, China
| |
Collapse
|
4
|
Spinner CD, Bell S, Einsele H, Tremblay C, Goldman M, Chagla Z, Finckh A, Edwards CJ, Aurer I, Launay O, Casañas I Comabella C, James S, Dube S, Borkowska K, Jah F, Kandeil W, Yokota RTC, Artaud C, Gottenberg JE, Gesualdo L, Bertrand D, Arnetorp S, Magiorkinis G. Is COVID-19 Still a Threat? An Expert Opinion Review on the Continued Healthcare Burden in Immunocompromised Individuals. Adv Ther 2024:10.1007/s12325-024-03043-0. [PMID: 39680311 DOI: 10.1007/s12325-024-03043-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/17/2024] [Indexed: 12/17/2024]
Abstract
Coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has had a profound global impact. The emergence of several variants during the pandemic has presented numerous challenges in preventing and managing this disease. The development of vaccines has played a pivotal role in controlling the pandemic, with a significant portion of the global population being vaccinated. This, along with the emergence of less virulent SARS-CoV-2 variants, has led to a reduction in the severity of COVID-19 outcomes for the overall population. Nevertheless, individuals with immunocompromising conditions continue to face challenges given their suboptimal response to vaccination and vulnerability to severe COVID-19. This expert review synthesizes recent published evidence regarding the economic and human impact of COVID-19 on such individuals. The literature suggests that rates of hospitalization, intensive care unit admission, and mechanical ventilation use were high during the pre-Omicron era, and remained high during Omicron and later, despite vaccination for this population. Moreover, studies indicated that these individuals experienced a negative impact on their mental health and health-related quality of life (HRQoL) compared to those without immunocompromising conditions, with elevated levels of anxiety, depression, and distress reported. Further, these individuals with immunocompromising conditions experienced substantial costs associated with COVID-19 and loss of income during the pandemic, though the evidence on the economic burden of COVID-19 in such individuals is limited. Generally, COVID-19 has increased healthcare resource use and costs, impaired mental health, and reduced HRQoL in those with varied immunocompromising conditions compared to both those without COVID-19 and the general population-underscoring the importance of continued real-world studies. Ongoing research is crucial to assess the ongoing burden of COVID-19 in vaccinated individuals with immunocompromising conditions who are still at risk of severe COVID-19 outcomes to ensure their needs are not disproportionately worse than the general population.
Collapse
Affiliation(s)
- Christoph D Spinner
- TUM School of Medicine and Health, Department of Clinical Medicine, Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, 81675, Munich, Germany.
| | - Samira Bell
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital, Dundee, Scotland
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Cécile Tremblay
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, H2X 0C1, Canada
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | | | | | | | - Christopher J Edwards
- NIHR Southampton Clinical Research Facility, University Hospital Southampton, NHS Foundation Trust, Southampton Hampshire, UK
| | - Igor Aurer
- University Hospital Center Zagreb Kišpatićeva ul. 12, and Medical School, University of Zagreb, Šalata 3, 10000, Zagreb, Croatia
| | - Odile Launay
- Université Paris Cité; Inserm CIC1417, F-CRIN I REIVAC; Assistance, Publique Hôpitaux de Paris, Cochin Hospital, Paris, France
| | | | - Samantha James
- Evidera, Evidence, Value and Access by PPD, Paris, France
| | - Sabada Dube
- Epidemiology, Vaccines and Immune Therapies Unit, AstraZeneca, Cambridge, UK
| | - Katarzyna Borkowska
- Evidera, Evidence, Value and Access by PPD, Granta Park Great Abington, Cambridge, UK
| | - Fungwe Jah
- Medical Affairs, BioPharmaceuticals Medical, AstraZeneca, Hamburg, Germany
| | - Walid Kandeil
- Vaccines and Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Baar, Switzerland
| | | | - Cécile Artaud
- Medical Affairs, Vaccines and Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Courbevoie, France
| | | | - Loreto Gesualdo
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Dominique Bertrand
- Department of Nephrology and Transplantation, University of Rouen, Rouen, France
| | - Sofie Arnetorp
- Health Economics & Payer Evidence, Vaccines & Immune Therapies, BioPharmaceuticals Medical, AstraZeneca, Gothenburg, Sweden
| | - Gkikas Magiorkinis
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 115 72, Athens, Greece
| |
Collapse
|
5
|
El-Zailik A, Sager J, Sánchez-Pearson Y, Parra S, Moore J, Sarkar P, Aylott A, Wang Q, Aldinger M, Garner C, Mogalian E, Skingsley A, Peppercorn A, Reyes M. Pharmacokinetics of the Monoclonal Antibody, Sotrovimab, in Healthy Participants Following IM Administration at Different Injection Sites. Clin Pharmacol Ther 2024. [PMID: 39668507 DOI: 10.1002/cpt.3515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024]
Abstract
Sotrovimab is a recombinant human monoclonal antibody for the early treatment of mild-to-moderate COVID- 19. A phase I, open-label, randomized, parallel-group study was conducted to investigate the pharmacokinetics, relative bioavailability, safety, and tolerability of two concentrations of sotrovimab administered intramuscularly at different injection sites in healthy volunteers. The study consisted of three parts (A, B, and C) and the pharmacokinetic results from Part A are reported herein. In Part A, participants were randomized in a 2:2:1:1 ratio to a 500 mg dose of 62.5 mg/mL sotrovimab administered into dorsogluteal muscle, or 100 mg/mL sotrovimab administered into dorsogluteal, anterolateral thigh, or deltoid muscles. Formulation concentration did not impact exposure following dorsogluteal administration; the point estimates (90% confidence interval [CI]) of the geometric mean ratios (GMRs) of area under the curve (AUC)inf and maximum serum concentration (Cmax) for dorsogluteal administration of 100 mg/mL vs. 62.5 mg/mL intramuscular sotrovimab were 0.95 (0.86-1.05) and 1.14 (1.02-1.27), respectively. However, the administration of 100 mg/mL sotrovimab in thigh or deltoid resulted in increased exposure relative to gluteal injections; the point estimates (90% CI) of the GMRs for 100 mg/mL intramuscular sotrovimab administered into thigh or deltoid muscles vs. 100 mg/mL administered dorsogluteally were 1.63 (1.46-1.83) and 1.50 (1.34-1.67) for AUCinf, and 1.82 (1.60-2.08) and 1.49 (1.31-1.69) for Cmax, respectively. Notably, thigh and deltoid administration also resulted in lower variability in key pharmacokinetic parameters such as AUC, Cmax, apparent clearance and volume of distribution, and earlier achievement of Cmax, than dorsogluteal intramuscular administration of sotrovimab.
Collapse
Affiliation(s)
| | | | | | - Sergio Parra
- Vir Biotechnology, Inc., San Francisco, California, USA
| | | | | | | | | | | | - Chad Garner
- Vir Biotechnology, Inc., San Francisco, California, USA
| | - Erik Mogalian
- Vir Biotechnology, Inc., San Francisco, California, USA
| | | | | | - Maribel Reyes
- Vir Biotechnology, Inc., San Francisco, California, USA
| |
Collapse
|
6
|
Huh K, Jo Y, Bae GH, Joo H, Radnaabaatar M, Lee H, Kim J, Kim DH, Yoo MG, Jo IU, Lee PH, Lee GW, Jung HS, Jung J. Nationwide Target Trial Emulation Evaluating the Clinical Effectiveness of Oral Antivirals for COVID-19 in Korea. J Korean Med Sci 2024; 39:e272. [PMID: 39497563 PMCID: PMC11538573 DOI: 10.3346/jkms.2024.39.e272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/30/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Despite the proven effectiveness of oral antivirals against severe acute respiratory syndrome coronavirus 2 in randomized trials, their clinical reevaluation is vital in the context of widespread immunity and milder prevalent variants. This study aimed to assess the effectiveness of oral antivirals for coronavirus disease 2019 (COVID-19). METHODS This retrospective cohort study utilized a target trial emulation framework to analyze patients with COVID-19 aged 60+ from January to December 2022. Data were obtained from the Korea Disease Control and Prevention Agency and Health Insurance Review and Assessment Service. The study involved 957,036 patients treated with nirmatrelvir/ritonavir and 243,360 treated with molnupiravir, each compared with the matched control groups. Primary outcome was progression to critical COVID-19 requiring advanced respiratory support. Secondary outcomes included progression to severe COVID-19, need for supplemental oxygen, and death within 30 days of the onset of COVID-19. Number needed to treat (NNT) derived from the absolute risk reduction. RESULTS Nirmatrelvir/ritonavir was significantly associated with a reduced risk of severe (adjusted odds ratio [aOR], 0.823; 95% confidence interval [CI], 0.803-0.843), critical (aOR, 0.560; 95% CI, 0.503-0.624), and fatal COVID-19 (aOR, 0.694; 95% CI, 0.647-0.744). Similarly, molnupiravir reduced the risk of severe (aOR, 0.895; 95% CI, 0.856-0.937), critical (aOR, 0.672; 95% CI, 0.559-0.807), and fatal cases (aOR, 0.679; 95% CI, 0.592-0.779). NNTs for nirmatrelvir/ritonavir were 203.71 (severe), 1,230.12 (critical), and 691.50 (death); for molnupiravir, they were 352.70 (severe), 1,398.62 (critical), and 862.98 (death). Higher effectiveness was associated with older adults, unvaccinated individuals, and the late pandemic phase. CONCLUSION Nirmatrelvir/ritonavir and molnupiravir are effective in preventing progression to severe disease in elderly adults with COVID-19.
Collapse
Affiliation(s)
- Kyungmin Huh
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Youngji Jo
- Department of Public Health Sciences, School of Medicine, University of Connecticut, Storrs, CT, USA
| | - Gi Hwan Bae
- Artificial Intelligence and Big-Data Convergence Center, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Hyejin Joo
- Department of Preventive Medicine, Gachon University College of Medicine, Incheon, Korea
| | - Munkhzul Radnaabaatar
- Artificial Intelligence and Big-Data Convergence Center, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Hyungmin Lee
- Patient Management Team, Central Headquarters of COVID-19, Korea Disease Control and Prevention Agency, Cheongju, Korea
- Division of Immunization, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Jungyeon Kim
- Patient Management Team, Central Headquarters of COVID-19, Korea Disease Control and Prevention Agency, Cheongju, Korea
- Division of Clinical Research, Center for Emerging Virus Research, National Institute of Infectious Disease, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Dong-Hwi Kim
- Patient Management Team, Central Headquarters of COVID-19, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Min-Gyu Yoo
- Patient Management Team, Central Headquarters of COVID-19, Korea Disease Control and Prevention Agency, Cheongju, Korea
- Division of Public Health Emergency Response Research, Bureau of Public Health Emergency Preparedness, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Il Uk Jo
- Big Data Management Division, Health Insurance Review & Assessment Service, Wonju, Korea
| | - Poong Hoon Lee
- Big Data Management Division, Health Insurance Review & Assessment Service, Wonju, Korea
| | - Geun Woo Lee
- Big Data Management Division, Health Insurance Review & Assessment Service, Wonju, Korea
| | - Hee Sun Jung
- Big Data Management Division, Health Insurance Review & Assessment Service, Wonju, Korea
| | - Jaehun Jung
- Artificial Intelligence and Big-Data Convergence Center, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
- Department of Preventive Medicine, Gachon University College of Medicine, Incheon, Korea.
| |
Collapse
|
7
|
Huang Y, Wang W, Liu Y, Wang Z, Cao B. COVID-19 vaccine updates for people under different conditions. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2323-2343. [PMID: 39083202 DOI: 10.1007/s11427-024-2643-1] [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: 04/19/2024] [Accepted: 06/06/2024] [Indexed: 10/22/2024]
Abstract
SARS-CoV-2 has caused global waves of infection since December 2019 and continues to persist today. The emergence of SARS-CoV-2 variants with strong immune evasion capabilities has compromised the effectiveness of existing vaccines against breakthrough infections. Therefore, it is important to determine the best utilization strategies for different demographic groups given the variety of vaccine options available. In this review, we will discuss the protective efficacy of vaccines during different stages of the epidemic and emphasize the importance of timely updates to target prevalent variants, which can significantly improve immune protection. While it is recognized that vaccine effectiveness may be lower in certain populations such as the elderly, individuals with chronic comorbidities (e.g., diabetes with poor blood glucose control, those on maintenance dialysis), or those who are immunocompromised compared to the general population, administering multiple doses can result in a strong protective immune response that outweighs potential risks. However, caution should be exercised when considering vaccines that might trigger an intense immune response in populations prone to inflammatory flare or other complications. In conclusion, individuals with special conditions require enhanced and more effective immunization strategies to prevent infection or reinfection, as well as to avoid the potential development of long COVID.
Collapse
Affiliation(s)
- Yijiao Huang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Weiyang Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yan Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- Department of Infectious Disease, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, 264000, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Bin Cao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China.
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
- Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing, 100084, China.
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Changping Laboratory, Beijing, 102200, China.
- Department of Respiratory Medicine, Capital Medical University, Beijing, 100069, China.
- New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
8
|
Reindl-Schwaighofer R, Heinzel A, Raab L, Strassl R, Herz CT, Regele F, Doberer K, Helk O, Spechtl P, Aschauer C, Hu K, Jagoditsch R, Reiskopf B, Böhmig GA, Benka B, Mahr B, Stiasny K, Weseslindtner L, Kammer M, Wekerle T, Oberbauer R. Cilgavimab and tixagevimab as pre-exposure prophylaxis in vaccine non-responder kidney transplant recipients during a period of prevalent SARS-CoV-2 BA.2 and BA.4/5 variants-a prospective cohort study (RESCUE-TX). EBioMedicine 2024; 109:105417. [PMID: 39442367 PMCID: PMC11539723 DOI: 10.1016/j.ebiom.2024.105417] [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: 06/03/2024] [Revised: 09/20/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND The response to severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) vaccination is severely impaired in patients on maintenance immunosuppression after kidney transplantation. METHODS We conducted a prospective cohort study of 194 kidney transplant recipients (KTR) who exhibited no response to SARS-CoV-2 vaccinations (i.e., SARS-CoV-2 spike protein antibodies ≤264 U/mL) and had no prior documented infection. Patients received 300 mg of cilgavimab/tixagevimab as SARS-CoV-2 pre-exposure prophylaxis (PrEP) between March 4, 2022, and May 3, 2022 and were contrasted to a matched cohort of 186 KTRs also without immunization again defined as SARS-CoV-2 spike protein antibodies ≤264 U/mL and no documented prior infection. The primary outcome was the serum kinetics of cilgavimab/tixagevimab, the secondary endpoints were time to SARS-CoV-2 breakthrough infection, severity of disease and variant specific live viral in vitro neutralization tests of patient sera. FINDINGS Longitudinal serum level monitoring showed a half-life of 91 days for both antibodies (95% CI 86-95 days for cilgavimab and 85-96 days for tixagevimab) in KTRs. In vitro neutralization tests showed effectiveness against the BA.2 omicron subvariant but not BA.5. The cumulative incidence of SARS-CoV-2 infections until May 15, 2022, (BA.2 dominance) was 15/194 vs 36/186 in the PrEP and control group respectively (OR = 0.35, 95% CI 0.18-0.66) but was not different thereafter (BA.4/5 dominance). The number of severe infections during the BA.2 period was lower in the prophylaxis than in the control group (OR = 0.37, 95% CI 0.17-0.79). INTERPRETATION This study showed that SARS-CoV-2 PrEP with cilgavimab/tixagevimab demonstrated clinical effectiveness against variants that are neutralised (BA.2) but not against BA.4/5. FUNDING This study was funded by the Medical University of Vienna and an unrestricted grant from AstraZeneca (ESR-21-21585).
Collapse
Affiliation(s)
- Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Heinzel
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lukas Raab
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Robert Strassl
- Department of Laboratory Medicine, Division of Clinical Virology, Medical University of Vienna, Vienna, Austria
| | - Carsten T Herz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Florina Regele
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Konstantin Doberer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Oliver Helk
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Paul Spechtl
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Constantin Aschauer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Karin Hu
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Rahel Jagoditsch
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Bianca Reiskopf
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Bernhard Benka
- Austrian Agency for Health and Food Safety (AGES), Vienna, Austria
| | | | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Michael Kammer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Center for Medical Data Science, Institute for Clinical Biometrics, Medical University of Vienna, Vienna, Austria
| | - Thomas Wekerle
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Rainer Oberbauer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Sung M, Kim YS, Cho C, Son Y, Kim DW, Lee SH. Impact of Immunosuppressants and Vaccination on COVID-19 Outcomes in Autoimmune Patients and Solid Organ Transplant Recipients: A Nationwide Propensity Score-Matched Study. Vaccines (Basel) 2024; 12:1190. [PMID: 39460355 PMCID: PMC11512354 DOI: 10.3390/vaccines12101190] [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: 09/19/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
PURPOSE This study investigates the impact of varying degrees of immunosuppression on the clinical outcomes of immunocompromised individuals, particularly those with autoimmune diseases or post-solid organ transplant statuses, in the context of COVID-19. By focusing on these highly vulnerable populations, the study underscores the significant health inequalities faced by immunocompromised patients, who experience disproportionately worse outcomes in comparison to the general population. METHODS A retrospective cohort analysis of the K-COV-N dataset was conducted, comparing the effects of immunosuppression in autoimmune and transplant groups with matched control groups. Propensity score matching was employed to minimize inequalities in baseline characteristics, ensuring a more equitable comparison between immunocompromised and non-immunocompromised individuals. Outcomes included COVID-19-related in-hospital mortality, 28-day mortality, ICU admissions, and the need for respiratory support among 323,890 adults in the Republic of Korea. Patients with cancer or other immunosuppressive conditions, such as HIV, were excluded. Subgroup analyses assessed the influence of specific immunosuppressive medications and vaccination extent. RESULTS Significantly elevated in-hospital mortality was found for patients with autoimmune diseases (adjusted Odds Ratio [aOR] 2.749) and transplant recipients (aOR 7.567), with similar patterns in other outcomes. High-dose steroid use and a greater number of immunosuppressant medications markedly increased the risk of poor outcomes. Vaccination emerged as a protective factor, with a single dose substantially improving outcomes for autoimmune patients and at least two doses necessary for transplant recipients. CONCLUSIONS Immunocompromised patients, particularly those with autoimmune diseases and transplant recipients, are highly vulnerable to severe COVID-19 outcomes. High-dose steroid use and multiple immunosuppressants further increase risks. Vaccination significantly improves outcomes, with at least one dose benefiting autoimmune patients and two doses necessary for transplant recipients. Personalized vaccination schedules based on immunosuppression levels are essential to mitigate healthcare inequalities and improve outcomes, particularly in underserved populations, informing both clinical and public health strategies.
Collapse
Affiliation(s)
- Mindong Sung
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.S.); (Y.-S.K.)
| | - Young-Sam Kim
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.S.); (Y.-S.K.)
| | - Changjin Cho
- Department of Information Statistics, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.C.); (Y.S.)
| | - Yongeun Son
- Department of Information Statistics, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.C.); (Y.S.)
| | - Dong-Wook Kim
- Department of Information Statistics, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.C.); (Y.S.)
- Department of Information and Statistics, Research Institute of Natural Science, Gyeongsang National University, 501 Jinju-daero, Jinju 52858, Republic of Korea
| | - Su-Hwan Lee
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.S.); (Y.-S.K.)
- Department of Bio & Medical Bigdata (BK21 Plus), Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
10
|
Boeckh M, Pergam SA, Limaye AP, Englund J, Corey L, Hill JA. How Immunocompromised Hosts Were Left Behind in the Quest to Control the COVID-19 Pandemic. Clin Infect Dis 2024; 79:1018-1023. [PMID: 38825885 PMCID: PMC11478583 DOI: 10.1093/cid/ciae308] [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] [Received: 03/20/2024] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024] Open
Abstract
The immunocompromised population was disproportionately affected by the severe acute respiratory syndrome coronavirus 2 pandemic. However, these individuals were largely excluded from clinical trials of vaccines, monoclonal antibodies, and small molecule antivirals. Although the community of scientists, clinical researchers, and funding agencies have proven that these therapeutics can be made and tested in record time, extending this progress to vulnerable and medically complex individuals from the start has been a missed opportunity. Here, we advocate that it is paramount to plan for future pandemics by investing in specific clinical trial infrastructure for the immunocompromised population to be prepared when the need arises.
Collapse
Affiliation(s)
- Michael Boeckh
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Steven A Pergam
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Ajit P Limaye
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Janet Englund
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
- Seattle Children's Research Institute, Seattle, Washington, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
Alirezaylavasani A, Skeie LG, Egner IM, Chopra A, Dahl TB, Prebensen C, Vaage JT, Halvorsen B, Lund-Johansen F, Tonby K, Reikvam DH, Stiksrud B, Holter JC, Dyrhol-Riise AM, Munthe LA, Kared H. Vaccine responses and hybrid immunity in people living with HIV after SARS-CoV-2 breakthrough infections. NPJ Vaccines 2024; 9:185. [PMID: 39384763 PMCID: PMC11464709 DOI: 10.1038/s41541-024-00972-3] [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: 02/26/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024] Open
Abstract
The COVID-19 pandemic posed a challenge for people living with HIV (PLWH), particularly immune non-responders (INR) with compromised CD4 T-cell reconstitution following antiretroviral therapy (CD4 count <350 cells per mm3). Their diminished vaccine responses raised concerns about their vulnerability to SARS-CoV-2 breakthrough infections (BTI). Our in-depth study here revealed chronic inflammation in PLWH and a limited anti-Spike IgG response after vaccination in INR. Nevertheless, the imprinting of Spike-specific B cells by vaccination significantly enhanced the humoral responses after BTI. Notably, the magnitude of cellular CD4 response in all PLWH was comparable to that in healthy donors (HD). However, the polyfunctionality and phenotype of Spike-specific CD8 T cells in INR differed from controls. The findings highlight the need for additional boosters with variant vaccines, and for monitoring ART adherence and the durability of both humoral and cellular anti-SARS-CoV-2 immunity in INR.
Collapse
Affiliation(s)
- Amin Alirezaylavasani
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Linda Gail Skeie
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Ingrid Marie Egner
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Adity Chopra
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Tuva Børresdatter Dahl
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Christian Prebensen
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - John Torgils Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dag Henrik Reikvam
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgitte Stiksrud
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Jan Cato Holter
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| |
Collapse
|
12
|
Im KI, Kim N, Lee J, Oh UH, Lee HW, Lee DG, Min GJ, Lee R, Lee J, Kim S, Cho SG. SARS-CoV-2-Specific T-Cell as a Potent Therapeutic Strategy against Immune Evasion of Emerging COVID-19 Variants. Int J Mol Sci 2024; 25:10512. [PMID: 39408840 PMCID: PMC11477143 DOI: 10.3390/ijms251910512] [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: 09/05/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Despite advances in vaccination and therapies for coronavirus disease, challenges remain due to reduced antibody longevity and the emergence of virulent variants like Omicron (BA.1) and its subvariants (BA.1.1, BA.2, BA.3, and BA.5). This study explored the potential of adoptive immunotherapy and harnessing the protective abilities using virus-specific T cells (VSTs). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) VSTs were generated by stimulating donor-derived peripheral blood mononuclear cells with spike, nucleocapsid, and membrane protein peptide mixtures. Phenotypic characterization, including T-cell receptor (TCR) vβ and pentamer analyses, was performed on the ex vivo-expanded cells. We infected human leukocyte antigen (HLA)-partially matched human Calu-3 cells with various authentic SARS-CoV-2 strains in a Biosafety Level 3 facility and co-cultured them with VSTs. VSTs exhibited a diverse TCR vβ repertoire, confirming their ability to target a broad range of SARS-CoV-2 antigens from both the ancestral and mutant strains, including Omicron BA.1 and BA.5. These ex vivo-expanded cells exhibited robust cytotoxicity and low alloreactivity against HLA-partially matched SARS-CoV-2-infected cells. Their cytotoxic effects were consistent across variants, targeting conserved spike and nucleocapsid epitopes. Our findings suggest that third-party partial HLA-matching VSTs could counter immune-escape mechanisms posed by emerging variants of concern.
Collapse
Affiliation(s)
- Keon-Il Im
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Nayoun Kim
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Junseok Lee
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
| | - Ui-Hyeon Oh
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Hye-Won Lee
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Dong-Gun Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.-G.L.); (R.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Gi-June Min
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Department of Hematology, Seoul St. Mary’s Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Raeseok Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.-G.L.); (R.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jinah Lee
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea; (J.L.); (S.K.)
| | - Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea; (J.L.); (S.K.)
| | - Seok-Goo Cho
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
- Department of Hematology, Seoul St. Mary’s Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
13
|
Calderaro DC, Valim V, Ferreira GA, Machado KLLL, Ribeiro PDC, Ribeiro SLE, Sartori NS, de Rezende RPV, de Melo AKG, Cruz VA, Vieira ASR, Kakehasi AM, de Landa AT, Burian APN, Peixoto FMMMC, Telles CMPF, do Espírito Santo RC, Baptista KL, de Oliveira YGP, Magalhães VDO, de Lima RL, Biegelmeyer E, Lorencini PZ, Teixeira-Carvalho A, dos Reis-Neto ET, Sato EI, Pinheiro MDM, Monticielo OA, de Souza VA, Xavier RM, Pileggi GS. Predictors of Hospitalization in Breakthrough COVID-19 among Fully Vaccinated Individuals with Immune-Mediated Rheumatic Diseases: Data from SAFER-Study. Vaccines (Basel) 2024; 12:1031. [PMID: 39340061 PMCID: PMC11436185 DOI: 10.3390/vaccines12091031] [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: 07/31/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Breakthrough COVID-19 (occurring in fully vaccinated people) has been described. Data on its characteristics among immune-mediated rheumatic disease (IMRD) patients are scarce. This study describes breakthrough COVID-19 occurring in IMRD patients participating in the SAFER-study, a Brazilian multicentric cohort evaluating the safety, effectiveness, and immunogenicity of SARS-CoV-2 vaccines in patients with autoimmune diseases. A descriptive analysis of the population and a binary logistic regression model were performed to evaluate the predictors of COVID-19-related hospitalization. A p-value < 0.05 was significant. The included 160 patients were predominantly females (83.1%), with a mean (SD) age of 40.23 (13.19) years. The patients received two (19%), three (70%), or four (11%) vaccine doses. The initial two-dose series was mainly with ChAdOx1 (Oxford/AstraZeneca) (58%) or BBIBP-CorV (Sinopharm-Beijing) (34%). The first booster (n = 150) was with BNT162b2 (BioNtech/Fosun Pharma/Pfizer) (63%) or ChAdOx1 (29%). The second booster (n = 112) was with BNT162b2 (40%) or ChAdOx1 (26%). The COVID-19 hospitalization rate was 17.5%. IMRD moderate/high activity (OR: 5.84; CI: 1.9-18.5; p = 0.002) and treatment with corticosteroids (OR: 2.94; CI: 1.02-8.49; p = 0.0043) were associated with higher odds of hospitalization, while increasing the number of vaccine doses was protective (OR: 0.37; CI: 0.15-0.9; p = 0.032). These findings, along with previous reassuring results about the safety of the COVID-19 vaccines, argue in favor of booster vaccination in IMRD patients.
Collapse
Affiliation(s)
- Débora Cerqueira Calderaro
- Locomotor System Department, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil; (G.A.F.)
| | - Valéria Valim
- Hospital Universitário Cassiano Antônio Moraes (HUCAM), Universidade Federal do Espírito Santo (UFES), Vitória 29041-295, ES, Brazil; (V.V.); (K.L.L.L.M.); (Y.G.P.d.O.); (P.Z.L.)
| | - Gilda Aparecida Ferreira
- Locomotor System Department, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil; (G.A.F.)
| | - Ketty Lysie Libardi Lira Machado
- Hospital Universitário Cassiano Antônio Moraes (HUCAM), Universidade Federal do Espírito Santo (UFES), Vitória 29041-295, ES, Brazil; (V.V.); (K.L.L.L.M.); (Y.G.P.d.O.); (P.Z.L.)
| | - Priscila Dias Cardoso Ribeiro
- Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil; (P.D.C.R.); (F.M.M.M.C.P.); (V.d.O.M.); (E.B.); (E.T.d.R.-N.); (E.I.S.); (M.d.M.P.); (G.S.P.)
| | - Sandra Lúcia Euzébio Ribeiro
- Escola de Medicina, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil; (S.L.E.R.); (C.M.P.F.T.); (R.L.d.L.)
| | - Natalia Sarzi Sartori
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90010-150, RS, Brazil; (N.S.S.); (R.C.d.E.S.); (O.A.M.); (R.M.X.)
| | | | - Ana Karla Guedes de Melo
- Hospital Universitário Lauro Wanderley, Universidade Federal da Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil;
| | - Vitor Alves Cruz
- Faculdade de Medicina, Universidade Federal de Goiás (UFG), Goiânia 74690-900, GO, Brazil;
| | | | - Adriana Maria Kakehasi
- Locomotor System Department, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil; (G.A.F.)
| | - Aline Teixeira de Landa
- Faculdade de Medicina, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil; (A.T.d.L.); (V.A.d.S.)
| | - Ana Paula Neves Burian
- Centro de Referências para Imunobiológicos Especiais (CRIE), Secretaria de Saúde do Estado do Espírito Santo, Vitória 29050-360, ES, Brazil;
| | - Flávia Maria Matos Melo Campos Peixoto
- Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil; (P.D.C.R.); (F.M.M.M.C.P.); (V.d.O.M.); (E.B.); (E.T.d.R.-N.); (E.I.S.); (M.d.M.P.); (G.S.P.)
| | | | - Rafaela Cavalheiro do Espírito Santo
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90010-150, RS, Brazil; (N.S.S.); (R.C.d.E.S.); (O.A.M.); (R.M.X.)
- Health Research and Innovation Science Centre, Klaipeda University, 92294 Klaipeda, Lithuania
| | - Katia Lino Baptista
- Universidade Federal Fluminense (UFF), Rio de Janeiro 24020-140, RJ, Brazil; (R.P.V.d.R.); (K.L.B.)
| | - Yasmin Gurtler Pinheiro de Oliveira
- Hospital Universitário Cassiano Antônio Moraes (HUCAM), Universidade Federal do Espírito Santo (UFES), Vitória 29041-295, ES, Brazil; (V.V.); (K.L.L.L.M.); (Y.G.P.d.O.); (P.Z.L.)
| | - Vanessa de Oliveira Magalhães
- Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil; (P.D.C.R.); (F.M.M.M.C.P.); (V.d.O.M.); (E.B.); (E.T.d.R.-N.); (E.I.S.); (M.d.M.P.); (G.S.P.)
| | - Raquel Lima de Lima
- Escola de Medicina, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil; (S.L.E.R.); (C.M.P.F.T.); (R.L.d.L.)
| | - Erika Biegelmeyer
- Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil; (P.D.C.R.); (F.M.M.M.C.P.); (V.d.O.M.); (E.B.); (E.T.d.R.-N.); (E.I.S.); (M.d.M.P.); (G.S.P.)
| | - Pietra Zava Lorencini
- Hospital Universitário Cassiano Antônio Moraes (HUCAM), Universidade Federal do Espírito Santo (UFES), Vitória 29041-295, ES, Brazil; (V.V.); (K.L.L.L.M.); (Y.G.P.d.O.); (P.Z.L.)
| | - Andréa Teixeira-Carvalho
- Instituto Renè Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte 30190-002, MG, Brazil;
| | - Edgard Torres dos Reis-Neto
- Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil; (P.D.C.R.); (F.M.M.M.C.P.); (V.d.O.M.); (E.B.); (E.T.d.R.-N.); (E.I.S.); (M.d.M.P.); (G.S.P.)
| | - Emília Inoue Sato
- Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil; (P.D.C.R.); (F.M.M.M.C.P.); (V.d.O.M.); (E.B.); (E.T.d.R.-N.); (E.I.S.); (M.d.M.P.); (G.S.P.)
| | - Marcelo de Medeiros Pinheiro
- Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil; (P.D.C.R.); (F.M.M.M.C.P.); (V.d.O.M.); (E.B.); (E.T.d.R.-N.); (E.I.S.); (M.d.M.P.); (G.S.P.)
| | - Odirlei André Monticielo
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90010-150, RS, Brazil; (N.S.S.); (R.C.d.E.S.); (O.A.M.); (R.M.X.)
| | - Viviane Angelina de Souza
- Faculdade de Medicina, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil; (A.T.d.L.); (V.A.d.S.)
| | - Ricardo Machado Xavier
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90010-150, RS, Brazil; (N.S.S.); (R.C.d.E.S.); (O.A.M.); (R.M.X.)
| | - Gecilmara Salviato Pileggi
- Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil; (P.D.C.R.); (F.M.M.M.C.P.); (V.d.O.M.); (E.B.); (E.T.d.R.-N.); (E.I.S.); (M.d.M.P.); (G.S.P.)
| |
Collapse
|
14
|
Benhamouda N, Besbes A, Bauer R, Mabrouk N, Gadouas G, Desaint C, Chevrier L, Lefebvre M, Radenne A, Roelens M, Parfait B, Weiskopf D, Sette A, Gruel N, Courbebaisse M, Appay V, Paul S, Gorochov G, Ropers J, Lebbah S, Lelievre JD, Johannes L, Ulmer J, Lebeaux D, Friedlander G, De Lamballerie X, Ravel P, Kieny MP, Batteux F, Durier C, Launay O, Tartour E. Cytokine profile of anti-spike CD4 +T cells predicts humoral and CD8 +T cell responses after anti-SARS-CoV-2 mRNA vaccination. iScience 2024; 27:110441. [PMID: 39104410 PMCID: PMC11298648 DOI: 10.1016/j.isci.2024.110441] [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: 04/08/2024] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Coordinating immune responses - humoral and cellular - is vital for protection against severe Covid-19. Our study evaluates a multicytokine CD4+T cell signature's predictive for post-vaccinal serological and CD8+T cell responses. A cytokine signature composed of four cytokines (IL-2, TNF-α, IP10, IL-9) excluding IFN-γ, and generated through machine learning, effectively predicted the CD8+T cell response following mRNA-1273 or BNT162b2 vaccine administration. Its applicability extends to murine vaccination models, encompassing diverse immunization routes (such as intranasal) and vaccine platforms (including adjuvanted proteins). Notably, we found correlation between CD4+T lymphocyte-produced IL-21 and the humoral response. Consequently, we propose a test that offers a rapid overview of integrated immune responses. This approach holds particular relevance for scenarios involving immunocompromised patients because they often have low cell counts (lymphopenia) or pandemics. This study also underscores the pivotal role of CD4+T cells during a vaccine response and highlights their value in vaccine immunomonitoring.
Collapse
Affiliation(s)
- Nadine Benhamouda
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Anissa Besbes
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | | | - Nesrine Mabrouk
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Gauthier Gadouas
- Bioinformatics and Cancer System Biology Team, IRCM-INSERM U1194, Institut de Recherche en Cancerologie de Montpellier, Montpellier, France
| | - Corinne Desaint
- INSERM SC10-US019, Villejuif, France
- Université Paris Cité, INSERM, CIC 1417, F-CRIN, Innovative Clinical Research Network in Vaccinology (I-REIVAC), APHP, CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Lucie Chevrier
- Université Paris Cité, INSERM U1016 Insitut Cochin, Hôpital Cochin, APHP, Centre Service d’immunologie Biologique, Paris, France
| | - Maeva Lefebvre
- Service de maladies infectieuses et tropicales, Centre de prévention des maladies infectieuses et transmissibles CHU de Nantes, Nantes, France
| | - Anne Radenne
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière-Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, APHP, Paris, France
| | - Marie Roelens
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Béatrice Parfait
- Centre de ressources Biologiques, Hôpital Cochin, APHP, Paris, France
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, School of Medicine in Health Sciences, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Nadège Gruel
- INSERM U830, Équipe Labellisée Ligue Nationale Contre le Cancer, Diversity and Plasticity of Childhood Tumors Lab, Centre de Recherche, Institut Curie, Université PSL, Paris, France
- Department of Translational Research, Centre de Recherche, Institut Curie, Université PSL, Paris, France
| | - Marie Courbebaisse
- Faculté de Médecine, Université Paris Cité, Paris, France
- Explorations fonctionnelles rénales, Physiologie, Hôpital Européen Georges-Pompidou, APHP, Paris, France
| | - Victor Appay
- Université de Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
- International Research Center of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Stephane Paul
- Centre International de Recherche en Infectiologie, Team GIMAP, Université Jean Monnet, Université Claude Bernard Lyon, INSERM, CIC 1408 INSERM Vaccinology, Immunology Department, iBiothera Reference Center, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Guy Gorochov
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses, APHP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jacques Ropers
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière –Hôpitaux Universitaires Pitié Salpêtrière- Charles Foix, APHP, Paris, France
| | - Said Lebbah
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière –Hôpitaux Universitaires Pitié Salpêtrière- Charles Foix, APHP, Paris, France
| | - Jean-Daniel Lelievre
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
- Groupe Henri-Mondor Albert-Chenevier, APHP, Créteil, France
| | - Ludger Johannes
- Cellular and Chemical Biology Unit, U1143 INSERM, UMR3666 CNRS, Institut Curie, Centre de Recherche, Université PSL, Paris, France
| | - Jonathan Ulmer
- Cellular and Chemical Biology Unit, U1143 INSERM, UMR3666 CNRS, Institut Curie, Centre de Recherche, Université PSL, Paris, France
| | - David Lebeaux
- Université Paris Cité, Service de maladies infectieuses Hôpital Saint Louis/Lariboisère APHP, INSERM, Paris, France
| | - Gerard Friedlander
- Department of « Croissance et Signalisation », Institut Necker Enfants Malades, INSERM U1151, CNRS UMR 8253, Université de Paris Cité, Paris, France
| | - Xavier De Lamballerie
- Unité des Virus Émergents, UVE: Aix-Marseille Université, IRD 190, INSERM 1207 Marseille, France
| | - Patrice Ravel
- Bioinformatics and Cancer System Biology Team, IRCM-INSERM U1194, Institut de Recherche en Cancerologie de Montpellier, Montpellier, France
| | - Marie Paule Kieny
- Institut National de la Santé et de la Recherche Médicale, INSERM, Paris, France
| | - Fréderic Batteux
- Université Paris Cité, INSERM U1016 Insitut Cochin, Hôpital Cochin, APHP, Centre Service d’immunologie Biologique, Paris, France
| | | | - Odile Launay
- Université Paris Cité, INSERM, CIC 1417, F-CRIN, Innovative Clinical Research Network in Vaccinology (I-REIVAC), APHP, CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Eric Tartour
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| |
Collapse
|
15
|
Shoham S. Convalescent Plasma for Immunocompromised Patients. Curr Top Microbiol Immunol 2024. [PMID: 39117848 DOI: 10.1007/82_2024_272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
COVID-19 convalescent plasma (CCP) is an important therapeutic option for immunocompromised patients with COVID-19. Such patients are at increased risk for serious complications of infection and may also develop a unique syndrome of persistent infection. This article reviews the rationale for CCP utilization in immunocompromised patients and the evidence for its value in immunosuppressed patients with both acute and persistent COVID-19. Both historical precedence and understanding of the mechanisms of action of antibody treatment support this use, as do several lines of evidence derived from case series, comparative studies, randomized trials, and systematic reviews of the literature. A summary of recommendations from multiple practice guidelines is also provided.
Collapse
Affiliation(s)
- Shmuel Shoham
- Department of Medicine, Johns Hopkins School of Medicine, 1830 East Monument St., Room 447, Baltimore, MD, 21205, USA.
| |
Collapse
|
16
|
Antequera A, Molin-Veglia AD, López-Alcalde J, Álvarez-Díaz N, Muriel A, Muñoz J. Reactivation of Trypanosoma cruzi infection in immunosuppressed patients: a systematic review and meta-analysis. Clin Microbiol Infect 2024; 30:980-988. [PMID: 38697392 DOI: 10.1016/j.cmi.2024.04.013] [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: 10/16/2023] [Revised: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND The risk of Trypanosoma cruzi reactivation is poorly understood. Previous studies evaluating the risk of reactivation report imprecise findings, and recommendations for monitoring and management from clinical guidelines rely on consensus opinion. OBJECTIVES We conducted a systematic review and meta-analysis to estimate the cumulative T. cruzi reactivation incidence in immunosuppressed adults, summarize the available evidence on prognostic factors for reactivation, and examine its prognostic effect on mortality. DATA SOURCES MEDLINE, Embase, LILACS, Clinical Trials, and CENTRAL from inception to 4 July 2022. STUDY ELIGIBILITY CRITERIA Studies reporting the incidence of T. cruzi reactivation. PARTICIPANTS Immunosuppressed adults chronically infected by T. cruzi. METHODS Two authors independently extracted data (including, but not limited to, incidence data, reactivation definition, follow-up, treatment, monitoring schedule, examined prognostic factors) and evaluated the risk of bias. We pooled cumulative incidence using a random-effects model. RESULTS Twenty-two studies (806 participants) were included. The overall pooled incidence of T. cruzi reactivation was 27% (95% CI, 19-36), with the highest pooled proportion in the sub-group of transplant recipients (36%; 95% CI, 25-48). The highest risk period was in the first 6 months after transplant (32%; 95% CI, 17-58), decreasing drastically the number of new cases later. People living with HIV and patients with autoimmune diseases experienced significantly lower cumulative reactivation incidences (17%; 95% CI, 8-29 and 18%; 95% CI, 9-29, respectively). A single study explored the independent effect of benznidazole and found benefits for preventing reactivations. No studies evaluated the independent association between reactivation and mortality, while sensitivity analysis results using unadjusted estimates were inconclusive. The heterogeneity of diagnostic algorithms was substantial. CONCLUSIONS Reactivation occurs in three out of ten T. cruzi-seropositive immunosuppressed adults. These findings can assist clinicians and panel guidelines in tailoring monitoring schedules. There is a great need for an accurate definition of reactivation and targeted monitoring.
Collapse
Affiliation(s)
- Alba Antequera
- Barcelona Institute for Global Health, Hospital Clínic Universitat de Barcelona (UB), Barcelona, Spain.
| | - Agustina Dal Molin-Veglia
- Barcelona Institute for Global Health, Hospital Clínic Universitat de Barcelona (UB), Barcelona, Spain
| | - Jesús López-Alcalde
- Faculty of Health Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain; Clinical Biostatistics Unit, Instituto Ramon y Cajal de Investigación Sanitaria, Madrid, Spain; CIBERESP, Madrid, Spain; Institute for Complementary and Integrative Medicine, University Hospital Zurich and University Zurich, Zurich, Switzerland
| | - Noelia Álvarez-Díaz
- Medical Library, Hospital Universitario Ramon y Cajal, Irycis, Madrid, Spain
| | - Alfonso Muriel
- Clinical Biostatistics Unit, Instituto Ramon y Cajal de Investigación Sanitaria, Madrid, Spain; CIBERESP, Madrid, Spain; Department of Nursing and Physiotherapy, Universidad Alcalá de Henares, Alcalá de Henares, Spain
| | - José Muñoz
- Barcelona Institute for Global Health, Hospital Clínic Universitat de Barcelona (UB), Barcelona, Spain
| |
Collapse
|
17
|
Montagud AC, Llenas-García J, Moragues R, Pérez-Bernabeu A, Alcocer Pertegal MJ, García Gómez FJ, Gamayo Serna AM, García Morante H, Caballero P, Tuells J. Counter reply in reference to "Prevalence of neutralizing antibodies against SARS-CoV-2 using a rapid serological test in health workers of a Spanish Department of Health in Alicante (Spain) before the booster dose of the vaccine". Rev Clin Esp 2024; 224:482-483. [PMID: 38852738 DOI: 10.1016/j.rceng.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Affiliation(s)
- A C Montagud
- Laboratorio de Inmunología, Plataforma Oncológica, Hospital QuironSalud Torrevieja, Alicante, Spain
| | - J Llenas-García
- Servicio de Medicina Interna, Hospital Vega Baja, Orihuela, Alicante, Spain; Departamento de Medicina Clínica, Universidad Miguel Hernández, Elche, Alicante, Spain; Fundación para la Promoción de la Salud e Investigación Biomédica de Valencia, FISABIO, Valencia, Spain
| | - R Moragues
- Departamento de Enfermería Comunitaria, Medicina Preventiva, Salud Pública e Historia de la Ciencia, Universidad de Alicante, Alicante, Spain
| | - A Pérez-Bernabeu
- Servicio de Medicina Interna, Hospital Vega Baja, Orihuela, Alicante, Spain; Fundación para la Promoción de la Salud e Investigación Biomédica de Valencia, FISABIO, Valencia, Spain
| | - M J Alcocer Pertegal
- Dirección de Enfermería de Atención Primaria, Departamento de Salud de Orihuela, Alicante, Spain
| | - F J García Gómez
- Dirección de Enfermería Hospitalaria, Hospital Vega Baja, Alicante, Spain
| | - A M Gamayo Serna
- Dirección de Enfermería Hospitalaria, Hospital Vega Baja, Alicante, Spain
| | - H García Morante
- Servicio de Medicina Interna, Hospital Vega Baja, Orihuela, Alicante, Spain; Fundación para la Promoción de la Salud e Investigación Biomédica de Valencia, FISABIO, Valencia, Spain
| | - P Caballero
- Departamento de Enfermería Comunitaria, Medicina Preventiva, Salud Pública e Historia de la Ciencia, Universidad de Alicante, Alicante, Spain
| | - J Tuells
- Departamento de Enfermería Comunitaria, Medicina Preventiva, Salud Pública e Historia de la Ciencia, Universidad de Alicante, Alicante, Spain; Instituto de Salud e Investigación Biomédica de Alicante, (ISABIAL), Alicante, Spain.
| |
Collapse
|
18
|
Hendy DA, Ma Y, Dixon TA, Murphy CT, Pena ES, Carlock MA, Ross TM, Bachelder EM, Ainslie KM, Fenton OS. Polymeric cGAMP microparticles affect the immunogenicity of a broadly active influenza mRNA lipid nanoparticle vaccine. J Control Release 2024; 372:168-175. [PMID: 38844178 PMCID: PMC11283345 DOI: 10.1016/j.jconrel.2024.06.007] [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: 12/14/2023] [Revised: 05/23/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024]
Abstract
Influenza outbreaks are a major burden worldwide annually. While seasonal vaccines do provide protection against infection, they are limited in that they need to be updated every year to account for the constantly mutating virus. Recently, lipid nanoparticles (LNPs) encapsulating mRNA have seen major success as a vaccine platform for SARS-CoV-2. Herein, we applied LNPs to deliver an mRNA encoding a computationally optimized broadly active (COBRA) influenza immunogen. These COBRA mRNA LNPs induced a broadly active neutralizing antibody response and protection after lethal influenza challenge. To further increase the immunogenicity of the COBRA mRNA LNPs, we combined them with acetalated dextran microparticles encapsulating a STING agonist. Contrary to recent findings, the STING agonist decreased the immunogenicity of the COBRA mRNA LNPs which was likely due to a decrease in mRNA translation as shown in vitro. Overall, this work aids in future selection of adjuvants to use with mRNA LNP vaccines.
Collapse
Affiliation(s)
- Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Timothy A Dixon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Connor T Murphy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Erik S Pena
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Michael A Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, USA
| | - Ted M Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, USA; Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA; Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA.
| |
Collapse
|
19
|
Chang HH, Lee YH, Huang KC, Chan DC, Lin YC, Sheng WH, Lee LT, Huang LM. COVID-19 vaccination: 2023 Taiwan Association of Gerontology and Geriatrics (TAGG) consensus statements. J Formos Med Assoc 2024:S0929-6646(24)00303-6. [PMID: 38991898 DOI: 10.1016/j.jfma.2024.06.027] [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: 05/28/2024] [Revised: 06/16/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024] Open
Abstract
The COVID-19 pandemic remains challenging due to the rapid evolution of the severe acute respiratory syndrome coronavirus 2. This article discusses recent findings on high-risk groups for COVID-19 mortality and morbidity, along with consensus statements from the 2023 Taiwan Association of Gerontology and Geriatrics (TAGG) meeting. It examines evidence on viral mutation mechanisms, emerging variants, and their implications for vaccination strategies. The article underscores advanced age, immunocompromised status, chronic medical conditions, occupational exposure, and socioeconomic disparities as significant risk factors for severe COVID-19 outcomes. TAGG's consensus emphasizes robust vaccination promotion, prioritizing elderly, and immunocompromised groups, individualized multi-dose regimens for immunocompromised patients, and simplified clinical guidelines. Discussions on global and regional recommendations for regular, variant-adapted boosters highlight the non-seasonal nature of COVID-19. Key agreements include escalating domestic preparedness, implementing vigorous risk-based vaccination, and adapting global guidelines to local contexts. Given ongoing viral evolution, proactive adjustment of vaccination policies is essential. Scientific consensus, tailored recommendations, and rapid knowledge dissemination are vital for optimizing COVID-19 protection among vulnerable groups in Taiwan. This article seeks to inform clinical practice and public health policy by summarizing expert-driven vaccination perspectives.
Collapse
Affiliation(s)
- Hao-Hsiang Chang
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Yi-Hsuan Lee
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Kuo-Chin Huang
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Ding-Cheng Chan
- Department of Geriatrics and Gerontology, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Ying-Chin Lin
- Geriatric Medicine Department, Taipei Medical University-Wanfang Hospital, Taipei, 116, Taiwan
| | - Wang-Huei Sheng
- College of Medicine, National Taiwan University, Taipei, 100, Taiwan; Department of Internal Medicine, National Taiwan University Children's Hospital, Taipei, 100, Taiwan
| | - Long-Teng Lee
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; College of Medicine, National Taiwan University, Taipei, 100, Taiwan; Taipei Jen-Chi Relief Institution, Taipei, 108, Taiwan.
| | - Li-Min Huang
- College of Medicine, National Taiwan University, Taipei, 100, Taiwan; Department of Pediatrics, National Taiwan University Children's Hospital, Taipei, 100, Taiwan.
| |
Collapse
|
20
|
Ko JH, Kim CM, Bang MS, Lee DY, Kim DY, Seo JW, Yun NR, Yang JY, Peck KR, Lee KW, Jung SH, Bang HJ, Bae WK, Kim TJ, Byeon KH, Kim SH, Kim DM. Risk Factors for Impaired Cellular or Humoral Immunity after Three Doses of SARS-CoV-2 Vaccine in Healthy and Immunocompromised Individuals. Vaccines (Basel) 2024; 12:752. [PMID: 39066390 PMCID: PMC11281526 DOI: 10.3390/vaccines12070752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Background: We aimed to identify the risk factors for impaired cellular and humoral immunity after three doses of the SARS-CoV-2 vaccine. Methods: Six months after the third vaccine dose, T-cell immunity was evaluated using interferon-gamma release assays (IGRAs) in 60 healthy and 139 immunocompromised (IC) individuals, including patients with hematologic malignancy (HM), solid malignancy (SM), rheumatic disease (RD), and kidney transplantation (KT). Neutralizing antibody titers were measured using the plaque reduction neutralization test (PRNT) and surrogate virus neutralization test (sVNT). Results: T-cell immunity results showed that the percentages of IGRA-positive results using wild-type/alpha spike protein (SP) and beta/gamma SP were 85% (51/60) and 75% (45/60), respectively, in healthy individuals and 45.6% (62/136) and 40.4% (55/136), respectively, in IC individuals. IC with SM or KT showed a high percentage of IGRA-negative results. The underlying disease poses a risk for impaired cellular immune response to wild-type SP. The risk was low when all doses were administered as mRNA vaccines. The risk factors for an impaired cellular immune response to beta/gamma SP were underlying disease and monocyte%. In the sVNT using wild-type SP, 12 of 191 (6.3%) individuals tested negative. In the PRNT of 46 random samples, 6 (13%) individuals tested negative for the wild-type virus, and 19 (41.3%) tested negative with omicrons. KT poses a risk for an impaired humoral immune response. Conclusions: Underlying disease poses a risk for impaired cellular immune response after the third dose of the SARS-CoV-2 vaccine; KT poses a risk for impaired humoral immune response, emphasizing the requirement of precautions in patients.
Collapse
Affiliation(s)
- Jae-Hoon Ko
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (J.-H.K.); (J.-Y.Y.); (K.-R.P.)
| | - Choon-Mee Kim
- Department of Premedical Science, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea;
| | - Mi-Seon Bang
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| | - Da-Yeon Lee
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| | - Da-Young Kim
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| | - Jun-Won Seo
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| | - Na-Ra Yun
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| | - Jin-Young Yang
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (J.-H.K.); (J.-Y.Y.); (K.-R.P.)
| | - Kyong-Ran Peck
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (J.-H.K.); (J.-Y.Y.); (K.-R.P.)
| | - Kyo-Won Lee
- Division of Transplantation, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
| | - Sung-Hoon Jung
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea; (S.-H.J.); (H.-J.B.); (W.-K.B.)
| | - Hyun-Jin Bang
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea; (S.-H.J.); (H.-J.B.); (W.-K.B.)
| | - Woo-Kyun Bae
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun 58128, Republic of Korea; (S.-H.J.); (H.-J.B.); (W.-K.B.)
| | - Tae-Jong Kim
- Department of Rheumatology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea;
| | - Kyeong-Hwan Byeon
- Department of Parasitology and Tropical Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea;
| | - Sung-Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
| | - Dong-Min Kim
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea; (M.-S.B.); (D.-Y.L.); (D.-Y.K.); (J.-W.S.); (N.-R.Y.)
| |
Collapse
|
21
|
Nasir N, Khanum I, Habib K, Wagley A, Arshad A, Majeed A. Insight into COVID-19 associated liver injury: Mechanisms, evaluation, and clinical implications. HEPATOLOGY FORUM 2024; 5:139-149. [PMID: 39006140 PMCID: PMC11237249 DOI: 10.14744/hf.2023.2023.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/25/2023] [Accepted: 11/02/2023] [Indexed: 07/16/2024]
Abstract
COVID-19 has affected millions worldwide, causing significant morbidity and mortality. While predominantly involving the respiratory tract, SARS-CoV-2 has also caused systemic illnesses involving other sites. Liver injury due to COVID-19 has been variably reported in observational studies. It has been postulated that liver damage may be due to direct damage by the SARS-CoV-2 virus or multifactorial secondary to hepatotoxic therapeutic options, as well as cytokine release syndrome and sepsis-induced multiorgan dysfunction. The approach to a COVID-19 patient with liver injury requires a thorough evaluation of the pattern of hepatocellular injury, along with the presence of underlying chronic liver disease and concurrent medications which may cause drug-induced liver injury. While studies have shown uneventful recovery in the majority of mildly affected patients, severe COVID-19 associated liver injury has been associated with higher mortality, prolonged hospitalization, and greater morbidity in survivors. Furthermore, its impact on long-term outcomes remains to be ascertained as recent studies report an association with metabolic-fatty liver disease. This present review provides insight into the subject by describing the postulated mechanism of liver injury, its impact in the presence of pre-existing liver disease, and its short- and long-term clinical implications.
Collapse
Affiliation(s)
- Nosheen Nasir
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Iffat Khanum
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Kiren Habib
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Abdullah Wagley
- Research Facilitation Office, Medical College, Aga Khan University, Karachi, Pakistan
| | - Aleena Arshad
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Atif Majeed
- Section of Gastroenterology, Department of Medicine, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
22
|
Theel ES, Kirby JE, Pollock NR. Testing for SARS-CoV-2: lessons learned and current use cases. Clin Microbiol Rev 2024; 37:e0007223. [PMID: 38488364 PMCID: PMC11237512 DOI: 10.1128/cmr.00072-23] [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] [Indexed: 06/14/2024] Open
Abstract
SUMMARYThe emergence and worldwide dissemination of SARS-CoV-2 required both urgent development of new diagnostic tests and expansion of diagnostic testing capacity on an unprecedented scale. The rapid evolution of technologies that allowed testing to move out of traditional laboratories and into point-of-care testing centers and the home transformed the diagnostic landscape. Four years later, with the end of the formal public health emergency but continued global circulation of the virus, it is important to take a fresh look at available SARS-CoV-2 testing technologies and consider how they should be used going forward. This review considers current use case scenarios for SARS-CoV-2 antigen, nucleic acid amplification, and immunologic tests, incorporating the latest evidence for analytical/clinical performance characteristics and advantages/limitations for each test type to inform current debates about how tests should or should not be used.
Collapse
Affiliation(s)
- Elitza S. Theel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - James E. Kirby
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Nira R. Pollock
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Chen DTH, Copland E, Hirst JA, Mi E, Dixon S, Coupland C, Hippisley-Cox J. Uptake, effectiveness and safety of COVID-19 vaccines in individuals at clinical risk due to immunosuppressive drug therapy or transplantation procedures: a population-based cohort study in England. BMC Med 2024; 22:237. [PMID: 38858672 PMCID: PMC11165729 DOI: 10.1186/s12916-024-03457-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 05/30/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Immunocompromised individuals are at increased risk of severe COVID-19 outcomes, underscoring the importance of COVID-19 vaccination in this population. The lack of comprehensive real-world data on vaccine uptake, effectiveness and safety in these individuals presents a critical knowledge gap, highlighting the urgency to better understand and address the unique challenges faced by immunocompromised individuals in the context of COVID-19 vaccination. METHODS We analysed data from 12,274,946 people in the UK aged > 12 years from 01/12/2020 to 11/04/2022. Of these, 583,541 (4.8%) were immunocompromised due to immunosuppressive drugs, organ transplants, dialysis or chemotherapy. We undertook a cohort analysis to determine COVID-19 vaccine uptake, nested case-control analyses adjusted for comorbidities and sociodemographic characteristics to determine effectiveness of vaccination against COVID-19 hospitalisation, ICU admission and death, and a self-controlled case series assessing vaccine safety for pre-specified adverse events of interest. RESULTS Overall, 93.7% of immunocompromised individuals received at least one COVID-19 vaccine dose, with 80.4% having received three or more doses. Uptake reduced with increasing deprivation (hazard ratio [HR] 0.78 [95%CI 0.77-0.79] in the most deprived quintile compared to the least deprived quintile for the first dose). Estimated vaccine effectiveness against COVID-19 hospitalisation 2-6 weeks after the second and third doses compared to unvaccinated was 78% (95%CI 72-83) and 91% (95%CI 88-93) in the immunocompromised population, versus 85% (95%CI 83-86) and 86% (95%CI 85-89), respectively, for the general population. Results showed COVID-19 vaccines were protective against intensive care unit (ICU) admission and death in both populations, with effectiveness of over 92% against COVID-19-related death and up to 95% in reducing ICU admissions for both populations following the third dose. COVID-19 vaccines were generally safe for immunocompromised individuals, though specific doses of ChAdOx1, mRNA-1273 and BNT162b2 raised risks of specific cardiovascular/neurological conditions. CONCLUSIONS COVID-19 vaccine uptake is high in immunocompromised individuals on immunosuppressive drug therapy or who have undergone transplantation procedures, with documented disparities by deprivation. Findings suggest that COVID-19 vaccines are protective against severe COVID-19 outcomes in this vulnerable population, and show a similar safety profile in immunocompromised individuals and the general population, despite some increased risk of adverse events. These results underscore the importance of ongoing vaccination prioritisation for this clinically at-risk population to maximise protection against severe COVID-19 outcomes.
Collapse
Affiliation(s)
- Daniel Tzu-Hsuan Chen
- Nuffield Department of Primary Care Health Science, University of Oxford, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - Emma Copland
- Nuffield Department of Primary Care Health Science, University of Oxford, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - Jennifer A Hirst
- Nuffield Department of Primary Care Health Science, University of Oxford, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - Emma Mi
- Nuffield Department of Primary Care Health Science, University of Oxford, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - Sharon Dixon
- Nuffield Department of Primary Care Health Science, University of Oxford, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - Carol Coupland
- Nuffield Department of Primary Care Health Science, University of Oxford, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
- Centre for Academic Primary Care, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Julia Hippisley-Cox
- Nuffield Department of Primary Care Health Science, University of Oxford, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK.
| |
Collapse
|
24
|
Pambrun E, Loubet P, Fourneron T, Moranne O. Immunogenicity of SARS-CoV-2 vaccines in patients treated with chronic double filtration plasmapheresis. J Clin Apher 2024; 39:e22136. [PMID: 38923591 DOI: 10.1002/jca.22136] [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: 01/02/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND The impact of chronic therapeutic plasmapheresis on humoral response following COVID-19 vaccination is poorly documented, especially among patients treated with double filtration plasmapheresis (DFPP). OBJECTIVES This retrospective single-center study evaluated the humoral response after SARS-CoV-2 vaccination and studied anti-SPIKE seropositivity and antibody dynamics in patients with chronic DFPP at our institution. METHOD All patients undergoing chronic DFPP at a tertiary center in France from December 2020 to November 2022 were included. We defined one patient subgroup as Group 1 to evaluate anti-SPIKE seropositivity after vaccination, with three groups based on their anti-SPIKE titers: (Group 1A) nonresponders (<0.8 UI/mL), (Group 1B) weak responders (0.8 to <250 binding antibody unit [BAU]/mL), and (Group 1C) strong responders (>250 BAU/mL). Group 2 served to evaluate antibody dynamics with anti-SPIKE levels measured 3 months after initial vaccination, Group 2A having a sustained level and Group 2B a declining pattern. RESULTS The 21 patients included had a median age of 63 years, and 13 (56%) were male. The indications for chronic DFPP mainly included dysimmune pathologies (15; 71%) and familial dyslipidemia (6; 29%). For the humoral response to vaccination in Patient Group 1, the only nonresponder was a patient who had undergone kidney transplantation 30 months earlier and was on immunosuppressive medication. For Patient Group 2, the median follow-up of antibody titers was 13 months [12-13]. Two distinct patterns of anti-SPIKE dynamics were observed: a rapid decline in anti-SPIKE antibody titers within 6 months following the initial vaccination or booster dose (n = 10 [71.4%] Group 2A) and stable anti-SPIKE levels above 250 BAU/mL over >6 months (n = 4 [28.6%] Group 2B) with more patients with familial dyslipidemia in the former. CONCLUSIONS Humoral response to SARS-CoV-2 vaccination appears robust in patients undergoing chronic DFPP and may be linked to patients' immune status rather than DFPTP itself. Our results support current recommendations for administering three doses of vaccine with a booster every 6 months.
Collapse
Affiliation(s)
- Emilie Pambrun
- Department of Nephrology-Dialysis-Apheresis, Nîmes University Hospital, Nîmes, France
| | - Paul Loubet
- Department of Infectious Disease, Nîmes University Hospital, Nîmes, France
| | - Thomas Fourneron
- Department of Nephrology-Dialysis-Apheresis, Nîmes University Hospital, Nîmes, France
| | - Olivier Moranne
- Department of Nephrology-Dialysis-Apheresis, Nîmes University Hospital, Nîmes, France
- IDESP, UMR-INSERM, Université de Montpellier, Montpellier, France
| |
Collapse
|
25
|
Zhang JR, Johnson JC, Preble RG, Mujtaba M, Lea AS, Stevenson HL, Kueht M. Beyond prevention: Unveiling the benefits of triple vaccination on COVID-19 severity and resource utilization in solid organ transplant recipients. Transpl Immunol 2024; 84:102048. [PMID: 38641149 DOI: 10.1016/j.trim.2024.102048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
OBJECTIVE Despite the widespread reduction in COVID-19-related morbidity and mortality attributed to vaccination in the general population, vaccine efficacy in solid organ transplant recipients (SOTR) remains under-characterized. This study aimed to investigate clinically relevant outcomes on double and triple-vaccinated versus unvaccinated SOTR with COVID-19. STUDY DESIGN AND SETTING A retrospective propensity score-matched cohort study was performed utilizing data from the US Collaborative Network Database within TriNetX (n = 117,905,631). We recruited vaccinated and unvaccinated (matched controls) SOTR with COVID-19 over two time periods to control for vaccine availability: December 2020 to October 2022 (bi-dose, double-dose vaccine effectiveness) and December 2020 to April 2023 (tri-dose, triple-dose vaccine effectiveness). A total of 42 factors associated with COVID-19 disease severity were controlled for including age, obesity, diabetes, and hypertension. We monitored 30-day outcomes including acute respiratory failure, intubation, and death following a diagnosis of COVID-19. RESULTS Subjects were categorized into two cohorts based on the two time periods: bi-dose cohort (vaccinated, n = 462; unvaccinated, n = 20,998); tri-dose cohort (vaccinated, n = 517; unvaccinated, n = 23,061).Compared to unvaccinated SOTR, 30-day mortality was significantly lower for vaccinated subjects in both cohorts: tri-dose (2.0% vs 7.5%, HR = 0.22 [95% CI: 0.11, 0.46]); bi-dose (3.7% vs 8.2%, HR = 0.43 [95% CI: 0.24, 0.76]). Hospital admission rates were similar between bi-dose vaccinated and unvaccinated subjects (33.1% vs 28.6%, HR = 1.2 [95% CI: 0.95, 1.52]). In contrast, tri-dose vaccinated subjects had a significantly lower likelihood of hospital admission (29.4% vs 36.6%, HR = 0.74 [95% CI: 0.6, 0.91]). Intubation rates were significantly lower for triple-vaccinated- (2.3% vs 5.2%, p < 0.05), but not double-vaccinated subjects (3.0% vs 5.2%, p > 0.05). CONCLUSION In solid organ transplant recipients with COVID-19, triple vaccination, but not double vaccination, against SARS-CoV-2 was associated with significantly less hospital resource utilization, decreased disease severity, and fewer short-term complications. These real-world data from extensively matched controls support the protective effects of COVID-19 vaccination with boosters in this vulnerable population.
Collapse
Affiliation(s)
- Jared R Zhang
- Department of Surgery, Division of Multiorgan Transplant and Hepatobiliary Surgery, University of Texas Medical Branch, Galveston, TX 77550, USA.
| | - John C Johnson
- Department of Surgery, Division of Multiorgan Transplant and Hepatobiliary Surgery, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Richard G Preble
- Department of Surgery, Division of Multiorgan Transplant and Hepatobiliary Surgery, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Muhammad Mujtaba
- Department of Medicine, Division of Transplant Nephrology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - A Scott Lea
- Department of Medicine, Division of Infectious Disease, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Heather L Stevenson
- Department of Pathology, Division of Transplant Pathology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Michael Kueht
- Department of Surgery, Division of Multiorgan Transplant and Hepatobiliary Surgery, University of Texas Medical Branch, Galveston, TX 77550, USA
| |
Collapse
|
26
|
Niessen FA, Bruijning-Verhagen PCJL, Bonten MJM, Knol MJ. Vaccine effectiveness against COVID-19 related hospital admission in the Netherlands by medical risk condition: A test-negative case-control study. Vaccine 2024; 42:3397-3403. [PMID: 38688804 DOI: 10.1016/j.vaccine.2024.04.017] [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: 09/26/2023] [Revised: 03/18/2024] [Accepted: 04/04/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Vaccination remains crucial in reducing COVID-19 hospitalizations and mitigating the strain on healthcare systems. We conducted a multicenter study to assess vaccine effectiveness (VE) of primary and booster vaccination against hospitalization and to identify subgroups with reduced VE. METHODS From March to July 2021 and October 2021 to January 2022, a test-negative case-control study was conducted in nine Dutch hospitals. The study included adults eligible for COVID-19 vaccination who were hospitalized with respiratory symptoms. Cases tested positive for SARS-CoV-2 within 14 days prior to or 48 h after admission, while controls tested negative. Logistic regression was used to calculate VE, adjusting for calendar week, sex, age, nursing home residency and comorbidity. We explored COVID-19 case characteristics and whether there are subgroups with less effective protection by vaccination against COVID-19 hospitalization. RESULTS Between October 2021 to January 2022, when the Delta variant was dominant, 335 cases and 277 controls were included. VE of primary and booster vaccination was 78 % (95 % CI: 65-86), and 89 % (95 % CI: 69-96), respectively. Using data from both study periods, including 700 cases and 511 controls, VE of primary vaccination was significantly reduced in those aged 60+ and patients with malignancy, chronic cardiac disease or an immunocompromising condition. CONCLUSION Although VE against hospitalization was 78% and increased to 89% after boosting during the Delta-dominant study period, VE was lower in certain high risk groups, for which indirect protection or other protective measures might be of added importance.
Collapse
Affiliation(s)
- F A Niessen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands.
| | - P C J L Bruijning-Verhagen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - M J M Bonten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - M J Knol
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| |
Collapse
|
27
|
Karunathilake RP, Kumara RA, Karunathilaka A, Wazil AWM, Nanayakkara N, Bandara CK, Abeysekera RA, Noordeen F, Gawarammana IB, Ratnatunga CN. 18-month longitudinal SARS COV-2 neutralizing antibody dynamics in haemodialysis patients receiving heterologous 3-dose vaccination (AZD-1222- AZD-1222- BNT162b2) in a lower middle income setting. BMC Nephrol 2024; 25:176. [PMID: 38778281 PMCID: PMC11112903 DOI: 10.1186/s12882-024-03599-7] [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: 12/06/2023] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Patients with chronic kidney disease on haemodialysis (HD) were given priority COVID-19 vaccination due to increased disease risk. The immune response to COVID-19 vaccination in patients on HD was diminished compared to healthy individuals in 2-dose studies. This study aimed to evaluate seroconversion rate, neutralizing antibody (nAB) levels and longitudinal antibody dynamics to 3-dose heterologous vaccination against COVID-19 in a cohort of HD patients compared to healthy controls and assess patient factors associated with antibody levels. METHODS This study was a case-control longitudinal evaluation of nAB dynamics in 74 HD patients compared to 37 healthy controls in a low/middle income setting. Corresponding samples were obtained from the two cohorts at time-points (TP) 1-1-month post 2nd dose of AZD1222 vaccine, TP2- 4 months post 2nd dose, TP4- 2 weeks post 3rd dose with BNT162b2 vaccine, TP5-5 months post 3rd dose and TP6-12 months post 3rd dose. Additional data is available at TP0- pre 2nd dose and TP3- 6 months post 2nd dose in HC and HD cohorts respectively. Anti-SARS-CoV-2 nAB were detected using Genscript cPassTM pseudoviral neutralization kit. Demographic and clinical details were obtained using an interviewer administered questionnaire. RESULTS Cohorts were gender matched while mean age of the HD cohort was 54.1yrs (vs HCs mean age, 42.6yrs, p < 0.05). Percentage seroconverted and mean/median antibody level (MAB) in the HD cohort vs HCs at each sampling point were, TP1-83.7% vs 100% (p < 0.05), MAB-450 IU/ml vs 1940 IU/ml (p < 0.0001); TP2-71.4% vs 100%, (p < 0.001), MAB- 235 IU/ml vs 453 IU/ml, (p < 0.05); TP4-95.2% vs 100% (p > 0.05), MAB-1029 IU/ml vs 1538 IU/ml (p < 0.0001); TP5-100% vs 100%, MAB-1542 IU/ml vs 1741IU/ml (p > 0.05); TP6-100% vs 100%, MAB-1961 IU/ml vs 2911 IU/ml (p > 0.05). At TP2, patients aged < 60 years (p < 0.001) were associated with maintaining seropositivity compared to patients > 60 years. CONCLUSION Two dose vaccination of haemodialysis patients provided poor nAB levels which improved markedly following 3rd dose vaccination, the effect of which was long- lasting with high nAB levels in both patients and controls detectable at 1 year follow-up.
Collapse
Affiliation(s)
| | - Roshan Athula Kumara
- Department of Microbiology, Faculty of Medicine, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | - Amali Karunathilaka
- Department of Microbiology, Faculty of Medicine, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | | | | | | | - Rajitha Asanga Abeysekera
- Department of Medicine, Faculty of Medicine, University of Peradeniya, Peradeniya, 20400, Sri Lanka
- Center for Education, Research and Training in Kidney Disease (CERTKiD), University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | - Faseeha Noordeen
- Department of Microbiology, Faculty of Medicine, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | | | | |
Collapse
|
28
|
Yaseen A, DeSantis SM, Sabharwal R, Talebi Y, Swartz MD, Zhang S, Leon Novelo L, Pinzon-Gomez CL, Messiah SE, Valerio-Shewmaker M, Kohl HW, Ross J, Lakey D, Shuford JA, Pont SJ, Boerwinkle E. Baseline characteristics of SARS-CoV-2 vaccine non-responders in a large population-based sample. PLoS One 2024; 19:e0303420. [PMID: 38739625 PMCID: PMC11090326 DOI: 10.1371/journal.pone.0303420] [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] [Received: 12/08/2023] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
INTRODUCTION Studies indicate that individuals with chronic conditions and specific baseline characteristics may not mount a robust humoral antibody response to SARS-CoV-2 vaccines. In this paper, we used data from the Texas Coronavirus Antibody REsponse Survey (Texas CARES), a longitudinal state-wide seroprevalence program that has enrolled more than 90,000 participants, to evaluate the role of chronic diseases as the potential risk factors of non-response to SARS-CoV-2 vaccines in a large epidemiologic cohort. METHODS A participant needed to complete an online survey and a blood draw to test for SARS-CoV-2 circulating plasma antibodies at four-time points spaced at least three months apart. Chronic disease predictors of vaccine non-response are evaluated using logistic regression with non-response as the outcome and each chronic disease + age as the predictors. RESULTS As of April 24, 2023, 18,240 participants met the inclusion criteria; 0.58% (N = 105) of these are non-responders. Adjusting for age, our results show that participants with self-reported immunocompromised status, kidney disease, cancer, and "other" non-specified comorbidity were 15.43, 5.11, 2.59, and 3.13 times more likely to fail to mount a complete response to a vaccine, respectively. Furthermore, having two or more chronic diseases doubled the prevalence of non-response. CONCLUSION Consistent with smaller targeted studies, a large epidemiologic cohort bears the same conclusion and demonstrates immunocompromised, cancer, kidney disease, and the number of diseases are associated with vaccine non-response. This study suggests that those individuals, with chronic diseases with the potential to affect their immune system response, may need increased doses or repeated doses of COVID-19 vaccines to develop a protective antibody level.
Collapse
Affiliation(s)
- Ashraf Yaseen
- The University of Texas Health Science Center at Houston, School of Public Health in Houston, Houston, TX, United States of America
| | - Stacia M. DeSantis
- The University of Texas Health Science Center at Houston, School of Public Health in Houston, Houston, TX, United States of America
| | - Rachit Sabharwal
- The University of Texas Health Science Center at Houston, School of Public Health in Houston, Houston, TX, United States of America
| | - Yashar Talebi
- The University of Texas Health Science Center at Houston, School of Public Health in Houston, Houston, TX, United States of America
| | - Michael D. Swartz
- The University of Texas Health Science Center at Houston, School of Public Health in Houston, Houston, TX, United States of America
| | - Shiming Zhang
- The University of Texas Health Science Center at Houston, School of Public Health in Houston, Houston, TX, United States of America
| | - Luis Leon Novelo
- The University of Texas Health Science Center at Houston, School of Public Health in Houston, Houston, TX, United States of America
| | - Cesar L. Pinzon-Gomez
- The University of Texas Health Science Center at Houston, School of Public Health in Houston, Houston, TX, United States of America
| | - Sarah E. Messiah
- The University of Texas Health Science Center at Houston, School of Public Health in Dallas, Dallas, TX, United States of America
- Center for Pediatric Population Health, UTHealth School of Public Health, Dallas, Texas, United States of America
| | - Melissa Valerio-Shewmaker
- The University of Texas Health Science Center at Houston, School of Public Health in Brownville, Brownsville, TX, United States of America
| | - Harold W. Kohl
- The University of Texas Health Science Center at Houston, School of Public Health in Austin, Austin, TX, United States of America
- University of Texas at Austin, Austin, TX, United States of America
| | - Jessica Ross
- The University of Texas Health Science Center at Houston, School of Public Health in Houston, Houston, TX, United States of America
| | - David Lakey
- University of Texas System, Austin, TX, United States of America
- The University of Texas Health Science Center Tyler, Tyler, TX, United States of America
| | - Jennifer A. Shuford
- Texas Department of State Health Services, Austin, TX, United States of America
| | - Stephen J. Pont
- Texas Department of State Health Services, Austin, TX, United States of America
| | - Eric Boerwinkle
- The University of Texas Health Science Center at Houston, School of Public Health in Houston, Houston, TX, United States of America
| |
Collapse
|
29
|
Thomas M, Masson M, Bitoun S, Hamroun S, Seror R, Dupuy H, Lazaro E, Richez C, Allanore Y, Avouac J. Prophylaxis with tixagevimab/cilgavimab is associated with lower COVID-19 incidence and severity in patients with autoimmune diseases. Rheumatology (Oxford) 2024; 63:1632-1638. [PMID: 37632774 DOI: 10.1093/rheumatology/kead449] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/28/2023] Open
Abstract
OBJECTIVE To describe the clinical efficacy of tixagevimab/cilgavimab in pre-exposure prophylaxis in patients at risk of severe coronavirus disease 2019 (COVID-19) and unresponsive to vaccination (anti-severe acute respiratory syndrome coronavirus 2 antibodies <260 binding antibody units/ml) in rheumatology. METHODS In this multicentre observational study we included patients with autoimmune or inflammatory diseases who received pre-exposure prophylaxis with tixagevimab/cilgavimab between December 2021 and August 2022. The endpoint was incidence of COVID-19 and its severity. RESULTS Tixagevimab/cilgavimab was administered to 115 patients with a median age of 62 years [interquartile range (IQR) 52-71], chronic arthritis (n = 53), connective tissue disease (n = 38) or vasculitis (n = 11). The main background immunosuppressants were rituximab (n = 98), corticosteroids [n = 62; median dose 5 mg (95% CI 5-8)] and methotrexate (n = 48). During a median follow-up of 128 days (IQR 93-173), COVID-19 occurred in 23/115 patients (20%) and the omicron variant was identified for the eight genotyped patients. During the study period, the average weekly incidence was 1071/100 000 inhabitants in Île-de-France vs 588/100 000 in our patients. Patients who received a two-injection regimen had a lower risk of infection than those with a single injection [16/49 (33%) vs 5/64 (8%), P = 0.0012]. The COVID-19-positive patients did not differ from uninfected patients concerning age, comorbidities, underlying rheumatic disease and immunosuppressants. All COVID-19 cases were non-severe. The tolerance of injections was excellent. CONCLUSION In a population with autoimmune or inflammatory diseases at risk of severe COVID-19 unresponsive to vaccination, pre-exposure prophylaxis withy tixagevimab/cilgavimab was associated with a lower incidence of COVID-19 and no severe infections.
Collapse
Affiliation(s)
- Marion Thomas
- Rheumatology Department, Cochin Hospital, Université Paris Cité, APHP, Paris, France
| | - Maeva Masson
- Rheumatology Department, Hôpital Purpan, Toulouse, France
| | - Samuel Bitoun
- Rheumatology Department, Hôpitaux Universitaires Paris-Sud, Le Kremlin Bicêtre, Paris, France
| | - Sabrina Hamroun
- Rheumatology Department, Cochin Hospital, Université Paris Cité, APHP, Paris, France
| | - Raphaele Seror
- Rheumatology Department, Hôpitaux Universitaires Paris-Sud, Le Kremlin Bicêtre, Paris, France
| | - Henry Dupuy
- Internal Medicine Department, Hôpitaux du Haut-Levêque, Pessac, France
| | - Estibaliz Lazaro
- Internal Medicine Department, Hôpitaux du Haut-Levêque, Pessac, France
| | - Christophe Richez
- Rheumatology Department, Centre Hospitalier Universitaire de Bordeaux-Groupe Hospitalier Pellegrin, Bordeaux, France
| | - Yannick Allanore
- Rheumatology Department, Cochin Hospital, Université Paris Cité, APHP, Paris, France
| | - Jérôme Avouac
- Rheumatology Department, Cochin Hospital, Université Paris Cité, APHP, Paris, France
| |
Collapse
|
30
|
Dalinkeviciene E, Gradauskiene B, Sakalauskaite S, Petruliene K, Vaiciuniene R, Skarupskiene I, Bastyte D, Sauseriene J, Valius L, Bumblyte IA, Ziginskiene E. Immune Response after Anti-SARS-CoV-2 mRNA Vaccination in Relation to Cellular Immunity, Vitamin D and Comorbidities in Hemodialysis Patients. Microorganisms 2024; 12:861. [PMID: 38792691 PMCID: PMC11123711 DOI: 10.3390/microorganisms12050861] [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: 03/11/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
In the global threat of SARS-CoV-2, individuals undergoing maintenance dialysis represent a vulnerable population with an increased risk of severe COVID-19 outcomes. Therefore, immunization against SARS-CoV-2 is an essential component of healthcare strategy for these patients. Existing data indicate that they tend to exhibit a reduced immune response to vaccines compared to the general population. Our study aimed to assess both humoral and cellular immune responses following two doses of an anti-SARS-CoV-2 mRNA vaccine, an ability to maintain adequate antibody titers over time, and potential relations with vitamin D, comorbidities and other factors in hemodialysis patients based on a single center experience. A total of 41/45 patients (91.1%) responded to the second dose of the anti-SARS-CoV-2 mRNA vaccine. The titer of anti-SARS-CoV-2 IgG class antibodies and levels of T cells three to four weeks after vaccination were lower in dialysis patients than in healthy controls. Antibodies titer in dialysis patients had a positive correlation with B lymphocytes and was related to cardiovascular diseases. The level of CD4+ cells had a negative correlation with hemodialysis vintage, as did the vitamin D level with post-vaccination seroconversion and decline in anti-SARS-CoV-2 antibodies titer during six months after vaccination. Hemodialysis patients had decreased amounts of CD4+ and CD8+ cells and lower levels of anti-SARS-CoV-2 antibodies than healthy controls. Therefore, chronic hemodialysis could lead to diminished cellular immunity and humoral immune response to the anti-SARS-CoV-2 mRNA vaccination and reduced protection from COVID-19. Comorbidity in cardiovascular diseases was associated with a lower level of specific anti-SARS-CoV-2 antibody titer. Vitamin D may be important in maintaining stable levels of anti-SARS-CoV-2 antibodies, while the duration of dialysis treatment could be one of the factors decreasing anti-SARS-CoV-2 antibody titer and determining lower CD4+ cell counts.
Collapse
Affiliation(s)
- Egle Dalinkeviciene
- Department of Nephrology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (K.P.); (R.V.); (I.S.); (I.A.B.); (E.Z.)
| | - Brigita Gradauskiene
- Department of Immunology and Allergology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania;
| | - Sandra Sakalauskaite
- Laboratory of Immunology, Department of Immunology and Allergology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (S.S.); (D.B.)
| | - Kristina Petruliene
- Department of Nephrology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (K.P.); (R.V.); (I.S.); (I.A.B.); (E.Z.)
| | - Ruta Vaiciuniene
- Department of Nephrology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (K.P.); (R.V.); (I.S.); (I.A.B.); (E.Z.)
| | - Inga Skarupskiene
- Department of Nephrology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (K.P.); (R.V.); (I.S.); (I.A.B.); (E.Z.)
| | - Daina Bastyte
- Laboratory of Immunology, Department of Immunology and Allergology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (S.S.); (D.B.)
| | - Jolanta Sauseriene
- Department of Family Medicine, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (J.S.); (L.V.)
| | - Leonas Valius
- Department of Family Medicine, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (J.S.); (L.V.)
| | - Inga Arune Bumblyte
- Department of Nephrology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (K.P.); (R.V.); (I.S.); (I.A.B.); (E.Z.)
| | - Edita Ziginskiene
- Department of Nephrology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (K.P.); (R.V.); (I.S.); (I.A.B.); (E.Z.)
| |
Collapse
|
31
|
Gazitt T, Eder L, Saliba W, Stein N, Feldhamer I, Cohen AD, Zisman D. COVID-19 Vaccine Effectiveness among Patients with Psoriatic Disease: A Population-Based Study. Vaccines (Basel) 2024; 12:453. [PMID: 38793704 PMCID: PMC11125670 DOI: 10.3390/vaccines12050453] [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: 03/01/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Limited information is available on the effectiveness of COVID-19 vaccination in patients with psoriasis and psoriatic arthritis (psoriatic disease (PsD)). The objective of our research was to assess the effectiveness of mRNA COVID-19 vaccination in preventing SARS-CoV-2 positivity and severe infection in a cohort of patients with PsD and the association of immunosuppressants on SARS-CoV-2 infection-related outcomes from December 2020 to December 2021. Vaccine effectiveness was assessed in a matched nested case control study using conditional logistic regression adjusted for demographics, comorbidities and immunosuppressant use. Study outcomes included SARS-CoV-2 positivity and severe COVID-19 (moderate-to-severe COVID-19-related hospitalizations or death). At least one dose of mRNA COVID-19 vaccine was associated with reduced risk of SARS-CoV-2 positivity and severe COVID-19 (OR = 0.41 (95% CI, 0.38-0.43) and OR = 0.15 (95% CI, 0.11-0.20), respectively). A more significant effect was found among patients who received three vaccines doses compared with those who did not receive any (OR (for positive SARS-CoV-2) = 0.13 (95% CI, 0.12-0.15) and OR (for severe disease) = 0.02 (0.01-0.05)). Etanercept and methotrexate were associated with higher risk of SARS-CoV-2 positivity (1.58 (1.19-2.10), p = 0.001 and 1.25 (1.03-1.51), p = 0.03, respectively). In conclusion, our results show that mRNA COVID-19 vaccines are effective in reducing both infection and severe COVID-19-related outcomes.
Collapse
Affiliation(s)
- Tal Gazitt
- Carmel Medical Center, Rheumatology Unit, Haifa 3436212, Israel;
- University of Washington Medical Center, Seattle, DC 98195, USA
- Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel;
| | - Lihi Eder
- Department of Medicine, Women’s College Hospital, University of Toronto, Toronto, ON M5S 3H2, Canada;
| | - Walid Saliba
- Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel;
- Department of Community Medicine and Epidemiology, Carmel Medical Center, Haifa 3436212, Israel;
| | - Nili Stein
- Department of Community Medicine and Epidemiology, Carmel Medical Center, Haifa 3436212, Israel;
| | - Ilan Feldhamer
- Chief Physician’s Office, Central Headquarters, Clalit Health Services, Tel Aviv 67754, Israel (A.D.C.)
| | - Arnon Dov Cohen
- Chief Physician’s Office, Central Headquarters, Clalit Health Services, Tel Aviv 67754, Israel (A.D.C.)
- Siaal Research Center for Family Medicine and Primary Care, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Devy Zisman
- Carmel Medical Center, Rheumatology Unit, Haifa 3436212, Israel;
- Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel;
| |
Collapse
|
32
|
Huh K, Kang M, Kim YE, Choi Y, An SJ, Seong J, Go MJ, Kang JM, Jung J. Risk of Severe COVID-19 and Protective Effectiveness of Vaccination Among Solid Organ Transplant Recipients. J Infect Dis 2024; 229:1026-1034. [PMID: 38097377 DOI: 10.1093/infdis/jiad501] [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: 06/14/2023] [Accepted: 11/13/2023] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Solid organ transplant recipients (SOTRs) are at higher risk for severe infection. However, the risk for severe COVID-19 and vaccine effectiveness among SOTRs remain unclear. METHODS This retrospective study used a nationwide health care claims database and COVID-19 registry from the Republic of Korea (2020 to 2022). Adult SOTRs diagnosed with COVID-19 were matched with up to 4 non-SOTR COVID-19 patients by propensity score. Severe COVID-19 was defined as treatment with high-flow nasal cannulae, mechanical ventilation, or extracorporeal membrane oxygenation. RESULTS Among 6783 SOTRs with COVID-19, severe COVID-19 was reported with the highest rate in lung transplant recipients (13.16%), followed by the heart (6.30%), kidney (3.90%), and liver (2.40%). SOTRs had a higher risk of severe COVID-19 compared to non-SOTRs, and lung transplant recipients showed the highest risk (adjusted odds ratio, 18.14; 95% confidence interval [CI], 8.53-38.58). Vaccine effectiveness against severe disease among SOTRs was 47% (95% CI, 18%-65%), 64% (95% CI, 49%-75%), and 64% (95% CI, 29%-81%) for 2, 3, and 4 doses, respectively. CONCLUSIONS SOTRs are at significantly higher risk for severe COVID-19 compared to non-SOTRs. Vaccination is effective in preventing the progression to severe COVID-19. Efforts should be made to improve vaccine uptake among SOTRs, while additional protective measures should be developed.
Collapse
Affiliation(s)
- Kyungmin Huh
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Minsun Kang
- Artificial Intelligence and Big-Data Convergence Center, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Young-Eun Kim
- Department of Bigdata Strategy, National Health Insurance Service, Wonju, South Korea
| | - Yoonkyung Choi
- Department of Bigdata Strategy, National Health Insurance Service, Wonju, South Korea
| | - Soo Jeong An
- Department of Big Data Management, National Health Insurance Service, Wonju, South Korea
| | - Jaehyun Seong
- Division of Clinical Research, Center for Emerging Virus Research, National Institute of Infectious Disease, National Institute of Health, Osong, South Korea
| | - Min Jin Go
- Division of Clinical Research, Center for Emerging Virus Research, National Institute of Infectious Disease, National Institute of Health, Osong, South Korea
| | - Ji-Man Kang
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, South Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Jaehun Jung
- Artificial Intelligence and Big-Data Convergence Center, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
- Department of Preventive Medicine, Gachon University College of Medicine, Incheon, South Korea
| |
Collapse
|
33
|
Arevalo-Romero JA, Chingaté-López SM, Camacho BA, Alméciga-Díaz CJ, Ramirez-Segura CA. Next-generation treatments: Immunotherapy and advanced therapies for COVID-19. Heliyon 2024; 10:e26423. [PMID: 38434363 PMCID: PMC10907543 DOI: 10.1016/j.heliyon.2024.e26423] [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: 10/12/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in 2019 following prior outbreaks of coronaviruses like SARS and MERS in recent decades, underscoring their high potential of infectivity in humans. Insights from previous outbreaks of SARS and MERS have played a significant role in developing effective strategies to mitigate the global impact of SARS-CoV-2. As of January 7, 2024, there have been 774,075,242 confirmed cases of COVID-19 worldwide. To date, 13.59 billion vaccine doses have been administered, and there have been 7,012,986 documented fatalities (https://www.who.int/) Despite significant progress in addressing the COVID-19 pandemic, the rapid evolution of SARS-CoV-2 challenges human defenses, presenting ongoing global challenges. The emergence of new SARS-CoV-2 lineages, shaped by mutation and recombination processes, has led to successive waves of infections. This scenario reveals the need for next-generation vaccines as a crucial requirement for ensuring ongoing protection against SARS-CoV-2. This demand calls for formulations that trigger a robust adaptive immune response without leading the acute inflammation linked with the infection. Key mutations detected in the Spike protein, a critical target for neutralizing antibodies and vaccine design -specifically within the Receptor Binding Domain region of Omicron variant lineages (B.1.1.529), currently dominant worldwide, have intensified concerns due to their association with immunity evasion from prior vaccinations and infections. As the world deals with this evolving threat, the narrative extends to the realm of emerging variants, each displaying new mutations with implications that remain largely misunderstood. Notably, the JN.1 Omicron lineage is gaining global prevalence, and early findings suggest it stands among the immune-evading variants, a characteristic attributed to its mutation L455S. Moreover, the detrimental consequences of the novel emergence of SARS-CoV-2 lineages bear a particularly critical impact on immunocompromised individuals and older adults. Immunocompromised individuals face challenges such as suboptimal responses to COVID-19 vaccines, rendering them more susceptible to severe disease. Similarly, older adults have an increased risk of severe disease and the presence of comorbid conditions, find themselves at a heightened vulnerability to develop COVID-19 disease. Thus, recognizing these intricate factors is crucial for effectively tailoring public health strategies to protect these vulnerable populations. In this context, this review aims to describe, analyze, and discuss the current progress of the next-generation treatments encompassing immunotherapeutic approaches and advanced therapies emerging as complements that will offer solutions to counter the disadvantages of the existing options. Preliminary outcomes show that these strategies target the virus and address the immunomodulatory responses associated with COVID-19. Furthermore, the capacity to promote tissue repair has been demonstrated, which can be particularly noteworthy for immunocompromised individuals who stand as vulnerable actors in the global landscape of coronavirus infections. The emerging next-generation treatments possess broader potential, offering protection against a wide range of variants and enhancing the ability to counter the impact of the constant evolution of the virus. Furthermore, advanced therapies are projected as potential treatment alternatives for managing Chronic Post-COVID-19 syndromeand addressing its associated long-term complications.
Collapse
Affiliation(s)
- Jenny Andrea Arevalo-Romero
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, 110231, Bogotá, D.C., Colombia
| | - Sandra M. Chingaté-López
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
| | - Bernardo Armando Camacho
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
| | - Carlos Javier Alméciga-Díaz
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, 110231, Bogotá, D.C., Colombia
| | - Cesar A. Ramirez-Segura
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
| |
Collapse
|
34
|
Titova E, Kan VW, Lozy T, Ip A, Shier K, Prakash VP, Starolis M, Ansari S, Goldgirsh K, Kim S, Pelliccia MC, Mccutchen A, Megalla M, Gunning TS, Kaufman HW, Meyer WA, Perlin DS. Humoral and cellular immune responses against SARS-CoV-2 post-vaccination in immunocompetent and immunocompromised cancer populations. Microbiol Spectr 2024; 12:e0205023. [PMID: 38353557 PMCID: PMC10913742 DOI: 10.1128/spectrum.02050-23] [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: 05/17/2023] [Accepted: 01/04/2024] [Indexed: 03/06/2024] Open
Abstract
Cancer patients are at risk for severe coronavirus disease 2019 (COVID-19) outcomes due to impaired immune responses. However, the immunogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination is inadequately characterized in this population. We hypothesized that cancer vs non-cancer individuals would mount less robust humoral and/or cellular vaccine-induced immune SARS-CoV-2 responses. Receptor binding domain (RBD) and SARS-CoV-2 spike protein antibody levels and T-cell responses were assessed in immunocompetent individuals with no underlying disorders (n = 479) and immunocompromised individuals (n = 115). All 594 individuals were vaccinated and of varying COVID-19 statuses (i.e., not known to have been infected, previously infected, or "Long-COVID"). Among immunocompromised individuals, 59% (n = 68) had an underlying hematologic malignancy; of those, 46% (n = 31) of individuals received cancer treatment <30 days prior to study blood collection. Ninety-eight percentage (n = 469) of immunocompetent and 81% (n = 93) of immunocompromised individuals had elevated RBD antibody titers (>1,000 U/mL), and of these, 60% (n = 281) and 44% (n = 41), respectively, also had elevated T-cell responses. Composite T-cell responses were higher in individuals previously infected with SARS-CoV-2 or those diagnosed with Long-COVID compared to uninfected individuals. T-cell responses varied between immunocompetent vs carcinoma (n = 12) cohorts (P < 0.01) but not in immunocompetent vs hematologic malignancy cohorts. Most SARS-CoV-2 vaccinated individuals mounted robust cellular and/or humoral responses, though higher immunogenicity was observed among the immunocompetent compared to cancer populations. The study suggests B-cell targeted therapies suppress antibody responses, but not T-cell responses, to SARS-CoV-2 vaccination. Thus, vaccination continues to be an effective way to induce humoral and cellular immune responses as a likely key preventive measure against infection and/or subsequent more severe adverse outcomes. IMPORTANCE The study was prompted by a desire to better assess the immune status of patients among our cancer host cohort, one of the largest in the New York metropolitan region. Hackensack Meridian Health is the largest healthcare system in New Jersey and cared for more than 75,000 coronavirus disease 2019 patients in its hospitals. The John Theurer Cancer Center sees more than 35,000 new cancer patients a year and performs more than 500 hematopoietic stem cell transplants. As a result, the work was undertaken to assess the effectiveness of vaccination in inducing humoral and cellular responses within this demographic.
Collapse
Affiliation(s)
- Elizabeth Titova
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Veronica W. Kan
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Tara Lozy
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Andrew Ip
- John Theurer Cancer Center, Hackensack, New Jersey, USA
- Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | | | | | | | - Sara Ansari
- Quest Diagnostics, Secaucus, New Jersey, USA
| | - Kira Goldgirsh
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Seoyeon Kim
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Michael C. Pelliccia
- John Theurer Cancer Center, Hackensack, New Jersey, USA
- Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Aamirah Mccutchen
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Martinus Megalla
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Thomas S. Gunning
- John Theurer Cancer Center, Hackensack, New Jersey, USA
- Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | | | | | - David S. Perlin
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| |
Collapse
|
35
|
Cossmann A, Hoffmann M, Stankov MV, Lürken K, Morillas Ramos G, Kempf A, Nehlmeier I, Pöhlmann S, Behrens GMN, Dopfer-Jablonka A. Immune responses following BNT162b2 XBB.1.5 vaccination in patients on haemodialysis in Germany. THE LANCET. INFECTIOUS DISEASES 2024; 24:e145-e146. [PMID: 38211602 DOI: 10.1016/s1473-3099(23)00783-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024]
Affiliation(s)
- Anne Cossmann
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Markus Hoffmann
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Metodi V Stankov
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Gema Morillas Ramos
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Amy Kempf
- Infection Biology Unit, German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Georg M N Behrens
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany; CiiM, Center for Individualised Infection Medicine, Hannover, Germany.
| | - Alexandra Dopfer-Jablonka
- Department of Rheumatology and Immunology, Hannover Medical School, 30625 Hannover, Germany; German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Hannover, Germany
| |
Collapse
|
36
|
Ju H, Seok JM, Chung YH, Jeon MY, Lee HL, Kwon S, Kim S, Min JH, Kim BJ. Evaluation of SARS-CoV-2 Vaccine-Induced Antibody Responses in Patients with Neuroimmunological Disorders: A Real-World Experience. Diagnostics (Basel) 2024; 14:502. [PMID: 38472974 DOI: 10.3390/diagnostics14050502] [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: 02/01/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
This study evaluates the antibody responses to SARS-CoV-2 vaccines in patients with neuroimmunological disorders (pwNID) who are receiving immunomodulating treatments, compared to healthy individuals. It included 25 pwNID with conditions such as optic neuritis, neuromyelitis optica spectrum disorder, multiple sclerosis, myasthenia gravis, and polymyositis, as well as 56 healthy controls. All participants had completed their full SARS-CoV-2 vaccination schedule, and their blood samples were collected within six months of their last dose. The concentration of anti-SARS-CoV-2 IgG antibodies was measured using an enzyme-linked immunosorbent assay. The results showed that pwNID had significantly lower antibody titers (58.4 ± 49.2 RU/mL) compared to healthy individuals (81.7 ± 47.3 RU/mL). This disparity persisted even after adjusting for age and the interval between the final vaccination and sample collection. A notable correlation was found between the use of immunomodulating treatments and reduced antibody levels, whereas mRNA vaccines were linked to higher antibody concentrations. The conclusion of this study is that immunomodulating treatments may reduce the effectiveness of SARS-CoV-2 vaccines in pwNID. This insight is crucial for healthcare providers in designing vaccination strategies and managing treatment plans for pwNID on immunomodulating therapies, highlighting the need for personalized approaches in this subgroup.
Collapse
Affiliation(s)
- Hyunjin Ju
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Department of Neurology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul 04401, Republic of Korea
| | - Jin Myoung Seok
- Department of Neurology, Soonchunhyang University Hospital Cheonan, Soonchunhyang University College of Medicine, Cheonan 31193, Republic of Korea
| | - Yeon Hak Chung
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Mi Young Jeon
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Hye Lim Lee
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Soonwook Kwon
- Department of Neurology, Inha University Hospital, Inha University College of Medicine, Incheon 22332, Republic of Korea
| | - Sunyoung Kim
- Department of Neurology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Ju-Hong Min
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Byoung Joon Kim
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| |
Collapse
|
37
|
Cavalera S, Di Nardo F, Serra T, Testa V, Baggiani C, Rosati S, Colitti B, Brienza L, Colasanto I, Nogarol C, Cosseddu D, Guiotto C, Anfossi L. A semi-quantitative visual lateral flow immunoassay for SARS-CoV-2 antibody detection for the follow-up of immune response to vaccination or recovery. J Mater Chem B 2024; 12:2139-2149. [PMID: 38315042 DOI: 10.1039/d3tb02895j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
The lateral flow immunoassay (LFIA) technique is largely employed for the point-of-care detection of antibodies especially for revealing the immune response in serum. Visual LFIAs usually provide the qualitative yes/no detection of antibodies, while quantification requires some equipment, making the assay more expensive and complicated. To achieve visual semi-quantification, the alignment of several lines (made of the same antigen) along a LFIA strip has been proposed. The numbering of the reacting lines has been used to correlate with the quantity of some biomarkers in serum. Here, we designed the first semiquantitative LFIA for detecting antibodies and applied it to classify the immune response to SARS-CoV-2 raised by vaccination or natural infection. We used a recombinant spike receptor-binding domain (RBD) as the specific capture reagent to draw two test lines. The detection reagent was selected among three possible ligands that are able to bind to anti-spike human antibodies: the same RBD, staphylococcal protein A, and anti-human immunoglobulin G antibodies. The most convenient detector, adsorbed on gold nanoparticles, was chosen based on the highest correlation with an antibody titre of 171 human sera, measured by a reference serological method, and was the RBD (Spearman's rho = 0.84). Incorporated into the semiquantitative LFIA, it confirmed the ability to discriminate high- and low-titre samples and to classify them into two classes (Dunn's test, P < 0.05). The proposed approach enabled the semiquantification of the immune response to SARS-CoV-2 by the unaided eye observation, thus overcoming the requirement of costly and complicated equipment, and represents a general strategy for the development of semiquantitative serological LFIAs.
Collapse
Affiliation(s)
- Simone Cavalera
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, Turin, Italy.
| | - Fabio Di Nardo
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, Turin, Italy.
| | - Thea Serra
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, Turin, Italy.
| | - Valentina Testa
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, Turin, Italy.
| | - Claudio Baggiani
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, Turin, Italy.
| | - Sergio Rosati
- Department of Veterinary Science, University of Turin, Largo Braccini 2, Grugliasco (TO), Italy
| | - Barbara Colitti
- Department of Veterinary Science, University of Turin, Largo Braccini 2, Grugliasco (TO), Italy
| | - Ludovica Brienza
- Department of Veterinary Science, University of Turin, Largo Braccini 2, Grugliasco (TO), Italy
| | - Irene Colasanto
- Department of Veterinary Science, University of Turin, Largo Braccini 2, Grugliasco (TO), Italy
| | - Chiara Nogarol
- In3diagnostic srl, Largo Braccini 2, Grugliasco (TO), Italy
| | - Domenico Cosseddu
- A.O. Ordine Mauriziano, Ospedale Umberto I di Torino, Via Magellano 1, Turin, Italy
| | - Cristina Guiotto
- A.O. Ordine Mauriziano, Ospedale Umberto I di Torino, Via Magellano 1, Turin, Italy
| | - Laura Anfossi
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, Turin, Italy.
| |
Collapse
|
38
|
Li R, Zhao JK, Li Q, Zhao L, Su YZ, Zhang JY, Zhang LY. Analysis of related factors for RA flares after SARS-CoV-2 infection: a retrospective study from patient survey. Sci Rep 2024; 14:4243. [PMID: 38378889 PMCID: PMC10879520 DOI: 10.1038/s41598-024-52748-3] [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: 07/03/2023] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
SARS-CoV-2 and its variants are widely prevalent worldwide. With frequent secondary and breakthrough infections, immune dysfunction in RA patients, and long-term use of immune preparations, SARS-CoV-2 infection poses a significant challenge to patients and rheumatologists. Whether SARS-CoV-2 infection causes RA flares and what factors aggravate RA flares are poorly studied. A questionnaire survey was conducted on RA patients infected with SARS-CoV-2 after December 7, 2022, in China through a multicenter and inter-network platform regarding general personal condition, primary disease, comorbidity, SARS-CoV-2 vaccination, viral infection, and impact on the primary disease. A total of 306 RA patients were included in this study, and the patient data were analyzed, in which the general condition of RA patients, medication use before SARS-CoV-2 infection and post-infection typing and manifestations, and medication adjustment did not affect the Flare of RA patients after SARS-CoV-2 infection. The control of disease before SARS-CoV-2 infection (OR = 2.10), RA involving pulmonary lesions (OR = 2.28), and the recovery time of COVID-19 (OR = 2.50) were risk factors for RA flare. RA involving pulmonary lesions, control status of disease before infection, and recovery time of COVID-19 disease are risk factors for RA flare after SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Rong Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Jun-Kang Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Qian Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Li Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ya-Zhen Su
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Jun-Yan Zhang
- Department of Clinical Epidemiology and Evidence-Based Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Li-Yun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
39
|
Anand ST, Vo AD, La J, Brophy M, Do NV, Fillmore NR, Branch-Elliman W, Monach PA. Risk of severe coronavirus disease 2019 despite vaccination in patients requiring treatment with immune-suppressive drugs: A nationwide cohort study of US Veterans. Transpl Infect Dis 2024; 26:e14168. [PMID: 37966134 DOI: 10.1111/tid.14168] [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: 07/20/2023] [Revised: 08/29/2023] [Accepted: 10/01/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Patients taking immune-suppressive drugs are at increased risk of severe coronavirus disease 2019 (COVID-19), not fully ameliorated by vaccination. We assessed the contributions of clinical and demographic factors to the risk of severe disease despite vaccination in patients taking immune-suppressive medications for solid organ transplantation (SOT), rheumatoid arthritis (RA), inflammatory bowel disease (IBD), or psoriasis. METHODS Veterans Health Administration electronic health records were used to identify patients diagnosed with RA, IBD, psoriasis, or SOT who had been vaccinated against severe acute respiratory syndrome coronavirus 2, were subsequently infected, and had received immune-suppressive drugs within 3 months before infection. The association of severe (defined as hypoxemia, mechanical ventilation, dexamethasone use, or death) versus non-severe COVID-19 with the use of immune-suppressive and antiviral drugs and clinical covariates was assessed by multivariable logistic regression. RESULTS Severe COVID-19 was more common in patients with SOT (230/1011, 22.7%) than RA (173/1355, 12.8%), IBD (51/742, 6.9%), or psoriasis (82/1125, 7.3%). Age was strongly associated with severe COVID-19, adjusted odds ratio (aOR) of 1.04 (CI 1.03-1.05) per year. Comorbidities indicating chronic brain, heart, lung, or kidney damage were also associated with severity, aOR 1.35-2.38. The use of glucocorticoids was associated with increased risk (aOR 1.66, CI 1.39-2.18). Treatment with antivirals was associated with reduced severity, for example, aOR 0.28 (CI 0.13-0.62) for nirmatrelvir/ritonavir. CONCLUSION The risk of severe COVID-19 despite vaccination is substantial in patients taking immune-suppressive drugs, more so in patients with SOT than in patients with inflammatory diseases. Age and severe comorbidities contribute to risk, as in the general population. Oral antivirals were very beneficial but not widely used.
Collapse
Affiliation(s)
- Sonia T Anand
- VA Boston Cooperative Studies Program, Boston, Massachusetts, USA
| | - Austin D Vo
- VA Boston Cooperative Studies Program, Boston, Massachusetts, USA
| | - Jennifer La
- VA Boston Cooperative Studies Program, Boston, Massachusetts, USA
| | - Mary Brophy
- VA Boston Cooperative Studies Program, Boston, Massachusetts, USA
- Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Nhan V Do
- VA Boston Cooperative Studies Program, Boston, Massachusetts, USA
- Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Nathanael R Fillmore
- VA Boston Cooperative Studies Program, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Westyn Branch-Elliman
- VA Boston Cooperative Studies Program, Boston, Massachusetts, USA
- Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- VA Boston Center for Healthcare Organization and Implementation Research, Boston, Massachusetts, USA
| | - Paul A Monach
- VA Boston Cooperative Studies Program, Boston, Massachusetts, USA
- Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Eribes E, Votruba C, Tinkham T, Huang A, Ilges D, Kunze K, Hudson M. Tixagevimab/cilgavimab for the prevention of COVID-19 in solid organ transplant recipients. Clin Transplant 2024; 38:e15261. [PMID: 38375915 DOI: 10.1111/ctr.15261] [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: 10/09/2023] [Revised: 12/27/2023] [Accepted: 01/29/2024] [Indexed: 02/21/2024]
Abstract
Tixagevimab/cilgavimab (tix/cil) received emergency use authorization in December 2021 for pre-exposure prophylaxis against COVID-19 in moderately to severely immunocompromised patients. Our study aimed to describe the incidence of COVID-19 infection and assess the immunologic risks associated with tix/cil in kidney, pancreas, liver, and heart transplant recipients. Retrospective chart review was completed to provide descriptive analysis. Outcomes data included COVID-19 infection, severity of COVID-19 infection, graft function, and rejection. Safety outcomes included cardiovascular (CV) and hypersensitivity events post tix/cil administration. A total of 410 transplant patients were included in the analysis: 20 heart, 92 liver, 243 kidney, 25 simultaneous pancreas/kidney, 23 simultaneous liver/kidney, and seven simultaneous heart/kidney. Twenty-seven (6.5%) patients tested positive for COVID-19 via PCR or antigen test post tix/cil. No apparent difference was observed in patients testing positive for COVID-19 by type of organ transplant (p = .122). Twenty-five of the 27 patients testing positive for COVID-19 reported symptomatic infection, only nine of whom were hospitalized. No patients were mechanically ventilated and no deaths due to COVID-19 occurred. No significant changes in graft function were observed. Clinically significant rejection was diagnosed and treated in four patients. COVID-19 breakthrough infection rates remained low in immunocompromised solid organ transplant recipients who received tix/cil. No significant immunologic risks were observed.
Collapse
Affiliation(s)
- Emily Eribes
- Department of Solid Organ Transplant, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Cassandra Votruba
- Department of Solid Organ Transplant, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Tyler Tinkham
- Department of Solid Organ Transplant, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Angela Huang
- Department of Solid Organ Transplant, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Dan Ilges
- Department of Solid Organ Transplant, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Katie Kunze
- Department of Solid Organ Transplant, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Madeline Hudson
- Department of Solid Organ Transplant, Mayo Clinic Arizona, Phoenix, Arizona, USA
| |
Collapse
|
41
|
Boretti A. mRNA vaccine boosters and impaired immune system response in immune compromised individuals: a narrative review. Clin Exp Med 2024; 24:23. [PMID: 38280109 PMCID: PMC10821957 DOI: 10.1007/s10238-023-01264-1] [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: 09/19/2023] [Accepted: 12/05/2023] [Indexed: 01/29/2024]
Abstract
Over the last 24 months, there has been growing evidence of a correlation between mRNA COVID-19 vaccine boosters and increased prevalence of COVID-19 infection and other pathologies. Recent works have added possible causation to correlation. mRNA vaccine boosters may impair immune system response in immune compromised individuals. Multiple doses of the mRNA COVID-19 vaccines may result in much higher levels of IgG 4 antibodies, or also impaired activation of CD4 + and CD8 + T cells. The opportunity for mRNA vaccine boosters to impair the immune system response needs careful consideration, as this impacts the cost-to-benefit ratio of the boosters' practice.
Collapse
Affiliation(s)
- Alberto Boretti
- Melbourne Institute of Technology, The Argus, 288 La Trobe St, Melbourne, VIC 3000, Australia.
| |
Collapse
|
42
|
Di Maira T, Vinaixa C, Izzy M, Paolo Russo F, Kirchner VA, Rammohan A, Belli LS, Polak WG, Berg T, Berenguer M. Worldwide variations in COVID-19 vaccination policies and practices in liver transplant settings: results of a multi-society global survey. FRONTIERS IN TRANSPLANTATION 2024; 2:1332616. [PMID: 38993892 PMCID: PMC11235330 DOI: 10.3389/frtra.2023.1332616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/15/2023] [Indexed: 07/13/2024]
Abstract
Background Despite the WHO's report of 24 available SARS-CoV-2 vaccines, limited data exist regarding vaccination policies for liver transplant (LT) patients. To address this, we conducted a global multi-society survey (EASL-ESOT-ELITA-ILTS) in LT centers. Methods A digital questionnaire assessing vaccine policies, safety, efficacy, and center data was administered online to LT centers. Results Out of 168 responding centers, 46.4%, 28%, 13.1%, 10.7%, and 1.8% were from European, American, Western Pacific, Southeast Asian, and Eastern Mediterranean Regions. Most LT centers prioritized COVID-19 vaccine access for LT patients (76%) and healthcare workers (86%), while other categories had lower priority (30%). One-third of responders recommended mRNA vaccine exclusively, while booster doses were widely recommended (81%). One-third conducted post-vaccine liver function tests post COVID-19 vaccine. Only 16% of centers modified immunosuppression, and mycophenolate discontinuation or modification was the main approach. Side effects were seen in 1 in 1,000 vaccinated patients, with thromboembolism, acute rejection, and allergic reaction being the most severe. mRNA showed fewer side effects (-3.1, p = 0.002). Conclusion COVID-19 vaccines and booster doses were widely used among LT recipients and healthcare workers, without a specific vaccine preference. Preventative immunosuppression adjustment post-vaccination was uncommon. mRNA vaccines demonstrated a favorable safety profile in this population.
Collapse
Affiliation(s)
- Tommaso Di Maira
- Liver Transplantation and Hepatology Unit, Hospital Universitari I Politècnic La Fe, Valencia, Spain
- Biomedical Research Network Center for Hepatic and Digestive Diseases (CIBEREHD), Madrid, Spain
- La Fe Health Research Institute, Valencia, Spain
| | - Carmen Vinaixa
- Liver Transplantation and Hepatology Unit, Hospital Universitari I Politècnic La Fe, Valencia, Spain
- Biomedical Research Network Center for Hepatic and Digestive Diseases (CIBEREHD), Madrid, Spain
- La Fe Health Research Institute, Valencia, Spain
| | - Manhal Izzy
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University Hospital Padua, Padua, Italy
| | - Varvara A. Kirchner
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA, United States
| | - Ashwin Rammohan
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, Bharath Institute of Higher Education and Research, Chennai, India
| | - Luca Saverio Belli
- Department of Hepatology and Gastroenterology, Niguarda Hospital, Milan, Italy
| | - Wojciech Grzegorz Polak
- Department of Surgery, Division of HPB and Transplant Surgery, Erasmus MC Transplant Institute, University Medical Centre, Rotterdam, Netherlands
| | - Thomas Berg
- Division of Hepatology, Department of Medicine II, Leipzig University, Medical Center, Leipzig, Germany
| | - Marina Berenguer
- Liver Transplantation and Hepatology Unit, Hospital Universitari I Politècnic La Fe, Valencia, Spain
- Biomedical Research Network Center for Hepatic and Digestive Diseases (CIBEREHD), Madrid, Spain
- La Fe Health Research Institute, Valencia, Spain
| |
Collapse
|
43
|
Priddey A, Chen-Xu MXH, Cooper DJ, MacMillan S, Meisl G, Xu CK, Hosmillo M, Goodfellow IG, Kollyfas R, Doffinger R, Bradley JR, Mohorianu II, Jones R, Knowles TPJ, Smith R, Kosmoliaptsis V. Microfluidic antibody profiling after repeated SARS-CoV-2 vaccination links antibody affinity and concentration to impaired immunity and variant escape in patients on anti-CD20 therapy. Front Immunol 2024; 14:1296148. [PMID: 38259440 PMCID: PMC10800570 DOI: 10.3389/fimmu.2023.1296148] [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: 09/18/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024] Open
Abstract
Background Patients with autoimmune/inflammatory conditions on anti-CD20 therapies, such as rituximab, have suboptimal humoral responses to vaccination and are vulnerable to poorer clinical outcomes following SARS-CoV-2 infection. We aimed to examine how the fundamental parameters of antibody responses, namely, affinity and concentration, shape the quality of humoral immunity after vaccination in these patients. Methods We performed in-depth antibody characterisation in sera collected 4 to 6 weeks after each of three vaccine doses to wild-type (WT) SARS-CoV-2 in rituximab-treated primary vasculitis patients (n = 14) using Luminex and pseudovirus neutralisation assays, whereas we used a novel microfluidic-based immunoassay to quantify polyclonal antibody affinity and concentration against both WT and Omicron (B.1.1.529) variants. We performed comparative antibody profiling at equivalent timepoints in healthy individuals after three antigenic exposures to WT SARS-CoV-2 (one infection and two vaccinations; n = 15) and in convalescent patients after WT SARS-CoV-2 infection (n = 30). Results Rituximab-treated patients had lower antibody levels and neutralisation titres against both WT and Omicron SARS-CoV-2 variants compared to healthy individuals. Neutralisation capacity was weaker against Omicron versus WT both in rituximab-treated patients and in healthy individuals. In the rituximab cohort, this was driven by lower antibody affinity against Omicron versus WT [median (range) KD: 21.6 (9.7-38.8) nM vs. 4.6 (2.3-44.8) nM, p = 0.0004]. By contrast, healthy individuals with hybrid immunity produced a broader antibody response, a subset of which recognised Omicron with higher affinity than antibodies in rituximab-treated patients [median (range) KD: 1.05 (0.45-1.84) nM vs. 20.25 (13.2-38.8) nM, p = 0.0002], underpinning the stronger serum neutralisation capacity against Omicron in the former group. Rituximab-treated patients had similar anti-WT antibody levels and neutralisation titres to unvaccinated convalescent individuals, despite two more exposures to SARS-CoV-2 antigen. Temporal profiling of the antibody response showed evidence of affinity maturation in healthy convalescent patients after a single SARS-CoV-2 infection, which was not observed in rituximab-treated patients, despite repeated vaccination. Discussion Our results enrich previous observations of impaired humoral immune responses to SARS-CoV-2 in rituximab-treated patients and highlight the significance of quantitative assessment of serum antibody affinity and concentration in monitoring anti-viral immunity, viral escape, and the evolution of the humoral response.
Collapse
Affiliation(s)
- Ashley Priddey
- Department of Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Michael Xin Hua Chen-Xu
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Daniel James Cooper
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Serena MacMillan
- Department of Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Catherine K. Xu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Myra Hosmillo
- Department of Pathology, Division of Virology, University of Cambridge, Cambridge, United Kingdom
| | - Ian G. Goodfellow
- Department of Pathology, Division of Virology, University of Cambridge, Cambridge, United Kingdom
| | - Rafael Kollyfas
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - John R. Bradley
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Irina I. Mohorianu
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
| | - Rachel Jones
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Tuomas P. J. Knowles
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, United Kingdom
| | - Rona Smith
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Vasilis Kosmoliaptsis
- Department of Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation at the University of Cambridge and the NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| |
Collapse
|
44
|
Moss J, Alexander L, Barré I, Parham I, Gillyard T, Davis J, Cunningham-Erves J. Understanding Physical Distancing and Face Mask Use Across High-Risk African American Subgroups During the COVID-19 Pandemic: Application of Health Belief Model. Health Promot Pract 2024; 25:49-59. [PMID: 36710489 PMCID: PMC9902790 DOI: 10.1177/15248399221151176] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Physical distancing and face masks remain frontline prevention strategies due to suboptimal vaccine uptake and the highly infectious COVID-19 variants. Communities of color are disproportionately impacted by a chronic disease burden that places them at higher risk of severe COVID-19 disease. Therefore, they can greatly benefit from face mask use and physical distancing, especially if the individual(s) have not received the vaccine. We applied the Health Belief Model to explore barriers and motivators influencing physical distancing and face mask use among high-risk, Black American subgroups during the early COVID-19 pandemic stages. We conducted 62 semi-structured interviews among four Black American subgroups: young adults, individuals with underlying medical conditions, essential workers, and parents. Thematic analysis, guided by the Health Belief Model, yielded six themes: (1) Knowledge on Face Mask Use and Physical Distancing, (2) Perceived Susceptibility and Severity Varies by Subgroup, (3) Experience with and Perceived Self-Efficacy to Engage in Preventive Behavior, (4) Perceived Benefits to engaging in preventive behaviors, (5) Perceived Barriers to engage in preventive behaviors, and (6) Cues to action to increase participation. Each subgroup's unique experience informed multilevel, tailored approaches that can be used by health promotion practitioners to improve face mask use and physical distancing among uniquely vulnerable Black American subgroups in the current and future pandemic.
Collapse
Affiliation(s)
- Jamal Moss
- Meharry Medical College, Nashville, TN, USA
| | | | - Iman Barré
- Meharry Medical College, Nashville, TN, USA
| | | | | | | | | |
Collapse
|
45
|
Ou S, Tancrède-Bohin E, Alexandre M, Ingen-Housz-Oro S, Castel M, Debarbieux S, Duvert-Lehembre S, Konstantinou MP, Abasq C, Berthin C, Couzan C, Lepelletier C, Jelti L, Bouteiller J, Calugareanu A, Véron M, Caux F, Joly P, Thomas B, Viguier M. Efficacy and safety of anti-COVID-19 vaccination in patients with autoimmune blistering diseases: A French national study. J Am Acad Dermatol 2024; 90:204-208. [PMID: 37769901 DOI: 10.1016/j.jaad.2023.08.101] [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] [Received: 04/26/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 10/03/2023]
Affiliation(s)
- Sokounthie Ou
- Department of Dermatology, Reims University Hospital, Centre de Référence des Maladies Bulleuses Auto-immunes, MALIBUL and EA7509, IRMAIC, Université Reims-Champagne Ardenne, Reims, France
| | - Emmanuelle Tancrède-Bohin
- Department of Dermatology, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Marina Alexandre
- Department of Dermatology, Groupe Hospitalier Paris Seine-Saint-Denis, Centre de Référence des Maladies Bulleuses Auto-immunes, MALIBUL and INSERM UMR1125, Bobigny, France
| | - Saskia Ingen-Housz-Oro
- Department of Dermatology, Henri-Mondor University Hospital, Assistance Publique - Hôpitaux de Paris, University Paris Est Créteil, EpidermE, Créteil, France
| | - Marion Castel
- Department of Dermatology, Rouen University Hospital, Centre de Référence des Maladies Bulleuses Auto-immunes, MALIBUL and INSERM U1234, Normandie University, Rouen, France
| | - Sébastien Debarbieux
- Department of Dermatology, Hospices Civils de Lyon, Lyon Sud Hospital, Lyon, France
| | - Sophie Duvert-Lehembre
- Department of Dermatology, Lille University Hospital, Lille, France; Department of Dermatology, Dunkerque Hospital, Dunkerque, France
| | | | - Claire Abasq
- Department of Dermatology, Augustin-Morvan Hospital, Brest, France
| | | | - Caroline Couzan
- Department of Dermatology, Saint-Étienne Hospital, Saint-Étienne, France
| | - Clémence Lepelletier
- Department of Dermatology, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Lamia Jelti
- Department of Dermatology, Henri-Mondor University Hospital, Assistance Publique - Hôpitaux de Paris, University Paris Est Créteil, EpidermE, Créteil, France
| | - Julie Bouteiller
- Department of Dermatology, Rouen University Hospital, Centre de Référence des Maladies Bulleuses Auto-immunes, MALIBUL and INSERM U1234, Normandie University, Rouen, France
| | | | - Marie Véron
- Department of Dermatology, Lille University Hospital, Lille, France
| | - Frédéric Caux
- Department of Dermatology, Groupe Hospitalier Paris Seine-Saint-Denis, Centre de Référence des Maladies Bulleuses Auto-immunes, MALIBUL and INSERM UMR1125, Bobigny, France
| | - Pascal Joly
- Department of Dermatology, Rouen University Hospital, Centre de Référence des Maladies Bulleuses Auto-immunes, MALIBUL and INSERM U1234, Normandie University, Rouen, France
| | - Bérenger Thomas
- Department of Epidemiology, Faculty of Medicine, Sorbonne University, Paris, France
| | - Manuelle Viguier
- Department of Dermatology, Reims University Hospital, Centre de Référence des Maladies Bulleuses Auto-immunes, MALIBUL and EA7509, IRMAIC, Université Reims-Champagne Ardenne, Reims, France.
| |
Collapse
|
46
|
Serra López-Matencio JM, Vicente-Rabaneda EF, Alañón E, Aranguren Oyarzabal A, Martínez Fleta P, Castañeda S. COVID-19 Vaccination and Immunosuppressive Therapy in Immune-Mediated Inflammatory Diseases. Vaccines (Basel) 2023; 11:1813. [PMID: 38140217 PMCID: PMC10747214 DOI: 10.3390/vaccines11121813] [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: 10/16/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
The COVID-19 vaccination program has probably been the most complex and extensive project in history until now, which has been a challenge for all the people involved in the planning and management of this program. Patients with immune-mediated inflammatory diseases (IMIDs) on immunosuppressive therapy have required special attention, not only because of the particular haste in carrying out the process but also because of the uncertainty regarding their response to the vaccines. We now have strong scientific evidence that supports the hypothesis that immunosuppressive therapy inhibits the humoral response to vaccines against other infectious agents, such as influenza, pneumococcus and hepatitis B. This has led to the hypothesis that the same could happen with the COVID-19 vaccine. Several studies have therefore already been carried out in this area, suggesting that temporarily discontinuing the administration of methotrexate for 2 weeks post-vaccination could improve the vaccine response, and other studies with various immunosuppressive drugs are in the same line. However, the fact of withholding or interrupting immunosuppressive therapy when dealing with COVID-19 vaccination remains unclear. On this basis, our article tries to compile the information available on the effect of immunosuppressant agents on COVID-19 vaccine responses in patients with IMIDs and proposes an algorithm for the management of these patients.
Collapse
Affiliation(s)
- José M. Serra López-Matencio
- Hospital Pharmacy Service, Hospital Universitario de La Princesa, IIS-Princesa, 28006 Madrid, Spain; (E.A.); (A.A.O.)
| | | | - Estefanía Alañón
- Hospital Pharmacy Service, Hospital Universitario de La Princesa, IIS-Princesa, 28006 Madrid, Spain; (E.A.); (A.A.O.)
| | - Ainhoa Aranguren Oyarzabal
- Hospital Pharmacy Service, Hospital Universitario de La Princesa, IIS-Princesa, 28006 Madrid, Spain; (E.A.); (A.A.O.)
| | - Pedro Martínez Fleta
- Immunology Service, Hospital Universitario de La Princesa, IIS-Princesa, 28006 Madrid, Spain;
| | - Santos Castañeda
- Rheumatology Service, Hospital Universitario de La Princesa, IIS-Princesa, 28006 Madrid, Spain;
| |
Collapse
|
47
|
Shirata M, Ito I, Tanaka M, Murata K, Murakami K, Ikeda H, Oi I, Hamao N, Nishioka K, Hayashi Y, Nagao M, Hashimoto M, Ito H, Ueno H, Morinobu A, Hirai T. Impact of methotrexate on humoral and cellular immune responses to SARS-CoV-2 mRNA vaccine in patients with rheumatoid arthritis. Clin Exp Med 2023; 23:4707-4720. [PMID: 37582911 DOI: 10.1007/s10238-023-01163-5] [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: 01/24/2023] [Accepted: 07/31/2023] [Indexed: 08/17/2023]
Abstract
The aim of this study was to longitudinally evaluate the undetermined impact of methotrexate (MTX) on the cumulative immunogenicity elicited by three doses of SARS-CoV-2 mRNA vaccination in patients with rheumatoid arthritis (RA). We prospectively evaluated vaccine-induced immune responses following the first dose, 1 and 6 months after the second dose, and 1 month after the third dose of BNT162b2 or mRNA-1273 in 144 SARS-CoV-2 naïve participants (70 patients with RA, 29 disease controls without immunosuppressive conditions, and 45 healthy controls). Humoral immune responses were assessed by quantifying anti-spike IgG antibody titers and the capacity of circulating antibodies to neutralize the ancestral SARS-CoV-2 strain and the Alpha, Delta, and Omicron variants. Vaccine-induced T-cell responses were measured using an interferon-gamma release assay. At 1 and 6 months after the second dose, anti-spike titers were highest in healthy controls, followed by disease controls and patients with RA. Multivariate analyses revealed that MTX treatment was significantly associated with a decrease in anti-spike titers, neutralizing activity, and SARS-CoV-2-specific interferon-gamma levels. Furthermore, MTX dose per body weight was negatively correlated with these two indices of humoral immune response. The third vaccine dose boosted anti-spike titers, especially in patients receiving MTX, while sera from these patients neutralized the Omicron variant far less robustly than those from healthy controls. In conclusion, MTX attenuated immunogenicity following two doses of SARS-CoV-2 mRNA vaccine in patients with RA, particularly resulting in dose-dependent suppression of the humoral immune response. Furthermore, MTX deteriorated the neutralizing activity against the Omicron variant, even after the third immunization.
Collapse
Affiliation(s)
- Masahiro Shirata
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
| | - Isao Ito
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan.
- Department of Internal Medicine, Sugita Genpaku Memorial Obama Municipal Hospital, Fukui, Japan.
| | - Masao Tanaka
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Murata
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kosaku Murakami
- Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Ikeda
- Department of Internal Medicine, Sugita Genpaku Memorial Obama Municipal Hospital, Fukui, Japan
| | - Issei Oi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
- Department of Internal Medicine, Sugita Genpaku Memorial Obama Municipal Hospital, Fukui, Japan
| | - Nobuyoshi Hamao
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
- Department of Internal Medicine, Sugita Genpaku Memorial Obama Municipal Hospital, Fukui, Japan
| | - Kensuke Nishioka
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
- Department of Internal Medicine, Sugita Genpaku Memorial Obama Municipal Hospital, Fukui, Japan
| | - Yasuyuki Hayashi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
| | - Miki Nagao
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motomu Hashimoto
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiromu Ito
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideki Ueno
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto, 606-8507, Japan
| |
Collapse
|
48
|
Russo C, Tagliafico L, Labate L, Ponzano M, Mirabella M, Portunato F, Bellezza C, Mora S, Arboscello E, Monacelli F, Nencioni A, Signori A, Bruzzone B, Giacomini M, Dentone C, Bassetti M. Effect of SARS-CoV-2 vaccination in a vulnerable COVID-19 cohort: a real-life experience in an Italian Hospital. J Chemother 2023; 35:730-736. [PMID: 37608747 DOI: 10.1080/1120009x.2023.2246716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 08/24/2023]
Abstract
Clinical trials demonstrated that SARS-CoV-2 vaccines reduce COVID-19-related mortality and morbidity. We describe the effect of vaccination on COVID-19-patients admitted at our hospital. Retrospective, single-center study conducted in Genoa, Italy, including patients ≥18years hospitalized for COVID-19 from May to December 2021. Demographical and clinical data were collected, vaccinated (group-A) and not-vaccinated (group-B) patients were compared. Impact of vaccination on mortality, ICU admission, and oxygen need was studied using Cox proportional hazards and logistic regression models after adjusting for propensity scores. Overall, 395 patients SARS-CoV-2 infected were included, of which 150 (38%) were vaccinated and 245 (62%) were not vaccinated. Patients in group-A were older, more disable, and with higher morbidity. Overall, 64 patients (16%) died within 30 days from admission, 34 in Group A (23%), and 30 in group B (12%). However, no statistically significant differences were observed (group-A versus group-B: HR 0.83, 95% CI 0.49-1.40, p = 0.483). On the other hand, vaccination was protective in terms of ICU admission (OR = 0.23, p = 0.046) and oxygen need (OR = 0.33, p = 0.008). Our study confirms that SARS-CoV-2 vaccination reduces morbidity among patients hospitalized for COVID-19. The still high mortality in our cohort of vaccinated individuals could be partially due to vulnerable conditions of our patients.
Collapse
Affiliation(s)
- Chiara Russo
- Division of Infectious Diseases, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Tagliafico
- Department of Internal Medicine (DIMI), Geriatric Clinic, University of Genoa, Genoa, Italy
| | - Laura Labate
- Division of Infectious Diseases, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Marta Ponzano
- Section of Biostatistics, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | | | | | - Carmen Bellezza
- Department of Emergency, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sara Mora
- Department of Informatics Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genoa, Italy
| | - Eleonora Arboscello
- Department of Emergency, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fiammetta Monacelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine (DIMI), Geriatric Clinic, University of Genoa, Genoa, Italy
| | - Alessio Nencioni
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine (DIMI), Geriatric Clinic, University of Genoa, Genoa, Italy
| | - Alessio Signori
- Section of Biostatistics, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Bianca Bruzzone
- Hygiene Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Mauro Giacomini
- Department of Informatics Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genoa, Italy
| | | | - Matteo Bassetti
- Division of Infectious Diseases, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
49
|
Benjakul S, Anthi AK, Kolderup A, Vaysburd M, Lode HE, Mallery D, Fossum E, Vikse EL, Albecka A, Ianevski A, Kainov D, Karlsen KF, Sakya SA, Nyquist-Andersen M, Gjølberg TT, Moe MC, Bjørås M, Sandlie I, James LC, Andersen JT. A pan-SARS-CoV-2-specific soluble angiotensin-converting enzyme 2-albumin fusion engineered for enhanced plasma half-life and needle-free mucosal delivery. PNAS NEXUS 2023; 2:pgad403. [PMID: 38077689 PMCID: PMC10703496 DOI: 10.1093/pnasnexus/pgad403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/13/2023] [Indexed: 02/29/2024]
Abstract
Immunocompromised patients often fail to raise protective vaccine-induced immunity against the global emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. Although monoclonal antibodies have been authorized for clinical use, most have lost their ability to potently neutralize the evolving Omicron subvariants. Thus, there is an urgent need for treatment strategies that can provide protection against these and emerging SARS-CoV-2 variants to prevent the development of severe coronavirus disease 2019. Here, we report on the design and characterization of a long-acting viral entry-blocking angiotensin-converting enzyme 2 (ACE2) dimeric fusion molecule. Specifically, a soluble truncated human dimeric ACE2 variant, engineered for improved binding to the receptor-binding domain of SARS-CoV-2, was fused with human albumin tailored for favorable engagement of the neonatal fragment crystallizable receptor (FcRn), which resulted in enhanced plasma half-life and allowed for needle-free transmucosal delivery upon nasal administration in human FcRn-expressing transgenic mice. Importantly, the dimeric ACE2-fused albumin demonstrated potent neutralization of SARS-CoV-2 immune escape variants.
Collapse
Affiliation(s)
- Sopisa Benjakul
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| | - Aina Karen Anthi
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| | - Anette Kolderup
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| | - Marina Vaysburd
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Heidrun Elisabeth Lode
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Department of Ophthalmology, Oslo University Hospital and University of Oslo, Oslo 0450, Norway
| | - Donna Mallery
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Even Fossum
- Department of Virology, Norwegian Institute of Public Health, Oslo 0213, Norway
| | - Elisabeth Lea Vikse
- Department of Virology, Norwegian Institute of Public Health, Oslo 0213, Norway
| | - Anna Albecka
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Denis Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
- Institute of Technology, University of Tartu, Tartu 50411, Estonia
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00290, Finland
| | - Karine Flem Karlsen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
| | - Siri Aastedatter Sakya
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| | - Mari Nyquist-Andersen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| | - Torleif Tollefsrud Gjølberg
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
- Department of Ophthalmology, Oslo University Hospital and University of Oslo, Oslo 0450, Norway
| | - Morten C Moe
- Department of Ophthalmology, Oslo University Hospital and University of Oslo, Oslo 0450, Norway
| | - Magnar Bjørås
- Department of Virology, Norwegian Institute of Public Health, Oslo 0213, Norway
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, Oslo 0371, Norway
| | - Leo C James
- Protein and Nucleic Acid Chemistry Division, Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Jan Terje Andersen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo 0372, Norway
| |
Collapse
|
50
|
Li Y, Zheng L, He H, Xiong H, Chen J, Sun H, Chen C, Li Q, Fu J, Wu F, Gao Y, Xian J, Liang M, Xiao G, Chen Q. First detection of cutavirus DNA in stools of patients with rheumatic diseases in Guangzhou, China. Virol Sin 2023; 38:860-867. [PMID: 37839551 PMCID: PMC10786651 DOI: 10.1016/j.virs.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023] Open
Abstract
Cutavirus (CuV) is a novel protoparvovirus possibly associated with diarrhea and cutaneous T-cell lymphomas. Patients with rheumatic disease are immunosuppressed and may be more vulnerable to pathogenic viruses. A descriptive study was conducted among hospitalized patients with rheumatic diseases and individuals undergoing medical health check-ups between June 2019 and June 2022 in Guangzhou, China. Stool samples of subjects were tested for CuV DNA. Demographic and fecal examination data of patients were obtained from electronic medical records. A total of 505 patients with rheumatic diseases and 244 individuals who underwent medical health check-ups were included in the study. Of the patients with rheumatic disease, 5.74% [95% confidence interval (CI): 4.03%-8.12%] were positive for CuV DNA, while no individual in the medical health check-up group was positive, indicating a close correlation between CuV and rheumatic disease. Men and patients with rheumatoid arthritis or ankylosing spondylitis, according to the disease classification, were more susceptible to being infected with CuV (P < 0.01). After adjustments, being male remained the only significant factor, with an adjusted odd ratio (OR) of 4.4 (95% CI: 1.7-11.4, P = 0.002). Phylogenetic analysis of the CuV VP2 sequences showed three diverse clades, one of which was segregated to be a single branching independent of previously known sequences, which is possible a new genotype.
Collapse
Affiliation(s)
- Yongzhi Li
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Liting Zheng
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Huan He
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Husheng Xiong
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jiaqi Chen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Hengbiao Sun
- Clinical Laboratory of Third Affiliated Hospital of Southern Medical University, Guangzhou, 510500, China
| | - Caiyun Chen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Qiushuang Li
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jiaqi Fu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Fei Wu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Yuhan Gao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Juxian Xian
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Minyi Liang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Gang Xiao
- Clinical Laboratory of Third Affiliated Hospital of Southern Medical University, Guangzhou, 510500, China.
| | - Qing Chen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|