1
|
Abba Moussa D, Vazquez M, Chable-Bessia C, Roux-Portalez V, Tamagnini E, Pedotti M, Simonelli L, Ngo G, Souchard M, Lyonnais S, Chentouf M, Gros N, Marsile-Medun S, Dinter H, Pugnière M, Martineau P, Varani L, Juan M, Calderon H, Naranjo-Gomez M, Pelegrin M. Discovery of a pan anti-SARS-CoV-2 monoclonal antibody with highly efficient infected cell killing capacity for novel immunotherapeutic approaches. Emerg Microbes Infect 2025; 14:2432345. [PMID: 39584380 PMCID: PMC11632933 DOI: 10.1080/22221751.2024.2432345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/24/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024]
Abstract
Unlocking the potential of broadly reactive coronavirus monoclonal antibodies (mAbs) and their derivatives offers a transformative therapeutic avenue against severe COVID-19, especially crucial for safeguarding high-risk populations. Novel mAb-based immunotherapies may help address the reduced efficacy of current vaccines and neutralizing mAbs caused by the emergence of variants of concern (VOCs). Using phage display technology, we discovered a pan-SARS-CoV-2 mAb (C10) that targets a conserved region within the receptor-binding domain (RBD) of the virus. Noteworthy, C10 demonstrates exceptional efficacy in recognizing all assessed VOCs, including recent Omicron variants. While C10 lacks direct neutralization capacity, it efficiently binds to infected lung epithelial cells and induces their lysis via natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity (ADCC). Building upon this pan-SARS-CoV-2 mAb, we engineered C10-based, Chimeric Antigen Receptor (CAR)-T cells endowed with efficient killing capacity against SARS-CoV-2-infected lung epithelial cells. Notably, NK and CAR-T-cell mediated killing of lung infected cells effectively reduces viral titers. These findings highlight the potential of non-neutralizing mAbs in providing immune protection against emerging infectious diseases. Our work reveals a pan-SARS-CoV-2 mAb effective in targeting infected cells and demonstrates the proof-of-concept for the potential application of CAR-T cell therapy in combating SARS-CoV-2 infections. Furthermore, it holds promise for the development of innovative antibody-based and cell-based therapeutic strategies against severe COVID-19 by expanding the array of therapeutic options available for high-risk populations.Trial registration: ClinicalTrials.gov identifier: NCT04093596.
Collapse
Affiliation(s)
| | - Mario Vazquez
- IDIBAPS, Immunogenetics and Immunotherapy in Autoinflammatory and Immune Responses, Barcelona, Spain
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Vincent Roux-Portalez
- IRCM, University of Montpellier, ICM, INSERM, Montpellier, France
- GenAc, Siric Plateform, INSERM, Montpellier, France
| | - Elia Tamagnini
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Mattia Pedotti
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Luca Simonelli
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Giang Ngo
- IRCM, University of Montpellier, ICM, INSERM, Montpellier, France
- PPM, BioCampus Plateforme de Protéomique de Montpellier CNRS, Montpellier, France
| | - Manon Souchard
- IRMB, University of Montpellier, INSERM, CNRS, Montpellier, France
| | | | - Myriam Chentouf
- IRCM, University of Montpellier, ICM, INSERM, Montpellier, France
- GenAc, Siric Plateform, INSERM, Montpellier, France
| | - Nathalie Gros
- CEMIPAI, University of Montpellier, UAR3725 CNRS, Montpellier, France
| | | | - Heiko Dinter
- IRMB, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Martine Pugnière
- IRCM, University of Montpellier, ICM, INSERM, Montpellier, France
- PPM, BioCampus Plateforme de Protéomique de Montpellier CNRS, Montpellier, France
| | - Pierre Martineau
- IRCM, University of Montpellier, ICM, INSERM, Montpellier, France
- GenAc, Siric Plateform, INSERM, Montpellier, France
| | - Luca Varani
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Manel Juan
- IDIBAPS, Immunogenetics and Immunotherapy in Autoinflammatory and Immune Responses, Barcelona, Spain
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Hugo Calderon
- IDIBAPS, Immunogenetics and Immunotherapy in Autoinflammatory and Immune Responses, Barcelona, Spain
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Mireia Pelegrin
- IRMB, University of Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
2
|
Body A, Lal L, Srihari S, MacIntyre CR, Buttery J, Ahern ES, Opat S, Leahy MF, Hamad N, Milch V, Turville S, Smith C, Lineburg K, Naing Z, Rawlinson W, Segelov E. Comprehensive humoral and cellular immune responses to COVID-19 vaccination in adults with cancer. Vaccine 2025; 46:126547. [PMID: 39648104 DOI: 10.1016/j.vaccine.2024.126547] [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: 09/07/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND The COVID-19 pandemic has significantly impacted people with cancer. Initial vaccine studies excluded patients with malignancy. Immunocompromised individuals remain vulnerable to SARS-CoV-2, necessitating detailed understanding of vaccine response. The epidemiology of COVID-19 in Australia offered unique opportunities to study cancer populations with minimal community exposure to SARS-CoV-2. METHODS SerOzNET prospectively examined previously unvaccinated patients with solid and haematological malignancies receiving up to five COVID-19 vaccine doses. Antibody response was measured by live virus neutralisation assay (neutralising antibody (NAb); positive titre ≥1:20; study primary endpoint) and commercial assay. T cell response was measured by cytometric bead array; positive defined as interferon gamma (IFN-γ) ≥10 pg/mL in response to Spike antigen. Patient and physician-reported adverse events were secondary endpoints. OUTCOMES 395 adults were enrolled prior to receiving mRNA vaccine (BNT162b2 = 347; mRNA-1273 = 1) or viral vector vaccine (ChadOx1-S = 43) for initial two-dose course, plus up to three additional doses. Median age was 58 years (range: 20-85); 60 % were female; 35 % had haematological malignancy, 2/395 (0.5 %) had baseline positive nucleocapsid antibody indicating prior SARS-CoV-2 exposure. NAb response post dose three was demonstrated in 84 % overall; 96 % of patients with solid cancers and 64 % with haematological cancer (p < 0·001). Risk factors for non-response were haematological cancer and anti B-cell therapies. Some patients with haematological cancer seroconverted for the first time after the fourth or fifth dose. IFN-γ response was seen in many patients with haematological cancer who lacked NAb response. Serious adverse events were rare. COVID-19 infection occurred in 29 % with no deaths. INTERPRETATION COVID-19 vaccination elicits B and T cell responses in patients with solid and haematological cancers, with an acceptable safety profile. A significant proportion of haematological cancer patients require >3 doses to elicit NAb, with many demonstrating T cell response, which may be an alternative pathway of immune protection.
Collapse
Affiliation(s)
- Amy Body
- Monash Health, Department of Oncology, Melbourne, VIC, Australia; Monash University, Department of Oncology, School of Clinical Sciences, Melbourne, VIC, Australia.
| | - Luxi Lal
- Monash Health, Department of Oncology, Melbourne, VIC, Australia; Monash University, Department of Oncology, School of Clinical Sciences, Melbourne, VIC, Australia
| | | | - C Raina MacIntyre
- Biosecurity Program, Kirby Institute, University of New South Wales, Sydney, NSW, Australia; School of Public Health and Community Medicine, University of New South Wales, Sydney, NSW, Australia; National Centre for Immunization, Research and Surveillance of Vaccine Preventable Diseases, University of Sydney, Westmead, NSW, Australia
| | - Jim Buttery
- University of Melbourne, Child Health Informatics (Paediatrics), Melbourne, VIC, Australia; Royal Children's Hospital, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Elizabeth Stephanie Ahern
- Monash Health, Department of Oncology, Melbourne, VIC, Australia; Monash University, Department of Oncology, School of Clinical Sciences, Melbourne, VIC, Australia
| | - Stephen Opat
- Monash Health, Department of Oncology, Melbourne, VIC, Australia; Monash University, Department of Oncology, School of Clinical Sciences, Melbourne, VIC, Australia
| | - Michael Francis Leahy
- Department of Haematology, Royal Perth Hospital, WA, Australia; University of Western Australia, School of Medicine & Pharmacology, School of Pathology, Perth, WA, Australia
| | - Nada Hamad
- Department of Haematology, St Vincent's Hospital, Kinghorn Cancer Centre, Sydney, NSW, Australia; The University of New South Wales, NSW, Australia
| | - Vivienne Milch
- Cancer Australia, Sydney, NSW, Australia; Caring Futures Institute, Flinders University, Adelaide, SA, Australia; School of Medicine, The University of Notre Dame Australia, Sydney, NSW, Australia
| | - Stuart Turville
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia; University of Sydney, NSW, Australia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; Queensland Immunology Research Centre, Brisbane, QLD, Australia
| | - Katie Lineburg
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Zin Naing
- Serology and Virology Division (SAViD), NSW Health Pathology East, Department of Microbiology, Prince of Wales Hospital, Randwick, Sydney, NSW, Australia
| | - William Rawlinson
- Serology and Virology Division (SAViD), NSW Health Pathology East, Department of Microbiology, Prince of Wales Hospital, Randwick, Sydney, NSW, Australia; Virology Research Laboratory, Prince of Wales Hospital, Randwick, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Eva Segelov
- Monash University, Department of Oncology, School of Clinical Sciences, Melbourne, VIC, Australia; University of Bern, Department of Clinical Research (Medicine), Bern, Switzerland; University Cancer Centre, Bern, Switzerland
| |
Collapse
|
3
|
Liang X, Yuan Y, Wang J, Tang C, Yang Y, Zhou Y, Yang H, Huang Q, Yu W, Wang H, Yan Y, Lin D, Li Y, Du X, Yuan L, Quan W, Wu D, Lu S. mRNA vaccines with RBD mutations have broad-spectrum activity against SARS-CoV-2 variants in mice. NPJ Vaccines 2025; 10:7. [PMID: 39805865 PMCID: PMC11729908 DOI: 10.1038/s41541-025-01066-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
The emergence of SARS-CoV-2 variants with defined mutations that enhance pathogenicity or facilitate immune evasion has resulted in a continual decline in the protective efficacy of existing vaccines. Therefore, there is a pressing need for a vaccine capable of combating future variants. In this study, we designed new mRNA vaccines, BSCoV05 and BSCoV06, and generated point mutations in the receptor-binding domain (RBD) of the original Wuhan strain to increase their broad-spectrum antiviral activity. Additionally, we used the BA.1 RBD as a control. Both vaccines elicited a robust immune response in BALB/c and K18-hACE2 mice, generating high levels of specific binding antibodies against the BA.2 RBD. Moreover, all three vaccines induced neutralizing antibodies against the prototype viral strain and relevant variants, including the Alpha and Beta strains and the Omicron variants BA.1, BA.2, BA.5, XBB.1.5, XBB.1.16, EG.5.1, and EG.5.1.1, with BSCoV06 demonstrating broader neutralizing antibody activity. Both BSCoV05 and BSCoV06 also elicited a cellular immune response. After the challenge, both BSCoV05 and BSCOV06 provided protection against the EG.5.1 strain in both mouse strains. Therefore, these two vaccines merit further evaluation in nonhuman primates, and this vaccine design strategy should be explored for its potential application in combating future SARS-CoV-2 variants, offering valuable insights into broad-spectrum vaccine development.
Collapse
Affiliation(s)
- Xiaoming Liang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Yuxia Yuan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Junbin Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Cong Tang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Yun Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Yanan Zhou
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Hao Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Qing Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Wenhai Yu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Haixuan Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Yuhuan Yan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Dongdong Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Yanwen Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Xuena Du
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Longhai Yuan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Wenqi Quan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Daoju Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China
| | - Shuaiyao Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical college, Kunming, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
- Yunnan Key Laboratory of Cross-Border Infectious Disease Control and Prevention and Novel Drug Development, Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Kunming, China.
| |
Collapse
|
4
|
Tyagi S, Tyagi N, Singh A, Gautam A, Singh A, Jindal S, Singh RP, Chaturvedi R, Kushwaha HR. Linking COVID-19 and cancer: Underlying mechanism. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167563. [PMID: 39510388 DOI: 10.1016/j.bbadis.2024.167563] [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: 03/25/2024] [Revised: 08/13/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), lead to a global health crisis with a spectrum of clinical manifestations. A potentially vulnerable category for SARS-CoV-2 infection was identified in patients with other medical conditions. Intriguingly, parallels exist between COVID-19 and cancer at the pathophysiological level, suggesting a possible connection between them. This review discusses all possible associations between COVID-19 and cancer. Expression of receptors like angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) increases COVID-19 susceptibility. SARS-CoV-2 infection might increase cancer susceptibility and accelerate cancer progression through mechanisms involving cytokine storm, tissue hypoxia, impaired T-cell responses, autophagy, neutrophil activation, and oxidative stress. These mechanisms collectively contribute to immune suppression, hindered apoptosis, and altered cellular signaling in the tumor microenvironment, creating conditions favorable for tumor growth, metastasis, and recurrence. Approved vaccines and their impact on cancer patients along-with new clinical trials are also described.
Collapse
Affiliation(s)
- Sourabh Tyagi
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Nipanshi Tyagi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Anu Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Akanksha Gautam
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Awantika Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Shelja Jindal
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rana P Singh
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India; School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Rupesh Chaturvedi
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India; School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Hemant Ritturaj Kushwaha
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India; School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
5
|
Mahrokhian SH, Tostanoski LH, Vidal SJ, Barouch DH. COVID-19 vaccines: Immune correlates and clinical outcomes. Hum Vaccin Immunother 2024; 20:2324549. [PMID: 38517241 PMCID: PMC10962618 DOI: 10.1080/21645515.2024.2324549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/24/2024] [Indexed: 03/23/2024] Open
Abstract
Severe disease due to COVID-19 has declined dramatically as a result of widespread vaccination and natural immunity in the population. With the emergence of SARS-CoV-2 variants that largely escape vaccine-elicited neutralizing antibody responses, the efficacy of the original vaccines has waned and has required vaccine updating and boosting. Nevertheless, hospitalizations and deaths due to COVID-19 have remained low. In this review, we summarize current knowledge of immune responses that contribute to population immunity and the mechanisms how vaccines attenuate COVID-19 disease severity.
Collapse
Affiliation(s)
- Shant H. Mahrokhian
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Lisa H. Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Samuel J. Vidal
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
6
|
Shapiro JR, Corrado M, Perry J, Watts TH, Bolotin S. The contributions of T cell-mediated immunity to protection from vaccine-preventable diseases: A primer. Hum Vaccin Immunother 2024; 20:2395679. [PMID: 39205626 PMCID: PMC11364080 DOI: 10.1080/21645515.2024.2395679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
In the face of the ever-present burden of emerging and reemerging infectious diseases, there is a growing need to comprehensively assess individual- and population-level immunity to vaccine-preventable diseases (VPDs). Many of these efforts, however, focus exclusively on antibody-mediated immunity, ignoring the role of T cells. Aimed at clinicians, public health practioners, and others who play central roles in human vaccine research but do not have formal training in immunology, we review how vaccines against infectious diseases elicit T cell responses, what types of vaccines elicit T cell responses, and how T cell responses are measured. We then use examples to demonstrate six ways that T cells contribute to protection from VPD, including directly mediating protection, enabling antibody responses, reducing disease severity, increasing cross-reactivity, improving durability, and protecting special populations. We conclude with a discussion of challenges and solutions to more widespread consideration of T cell responses in clinical vaccinology.
Collapse
Affiliation(s)
- Janna R. Shapiro
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Mario Corrado
- Division of General Internal Medicine, University of Toronto, Toronto, ON, Canada
| | - Julie Perry
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Tania H. Watts
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Shelly Bolotin
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Health Protection, Public Health Ontario, Toronto, ON, Canada
| |
Collapse
|
7
|
Yoshida M, Taguchi N, Piao Y, Gupta R, Peters J, Abdelghany M, Chiang M, Wang CY, Berry M, Yotsuyanagi H. Treatment patterns and clinical outcomes of immunocompromised patients with COVID-19 receiving remdesivir in the inpatient setting in Japan. J Infect Chemother 2024; 31:102578. [PMID: 39647701 DOI: 10.1016/j.jiac.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
INTRODUCTION Remdesivir (RDV) was approved in Japan for the treatment of coronavirus disease 2019 (COVID-19) in May 2020. However, large-scale research describing the characterized use of RDV in the inpatient setting in Japan is limited. This study aimed to describe the treatment patterns and clinical outcomes of immunocompromised patients with COVID-19 treated with RDV. METHODS We used a secondary nationwide administrative claims database of acute care hospitals in Japan. The study period was from October 19, 2020, to September 30, 2022. Patients with COVID-19 treated with RDV during hospitalization were included, and the proportion of patients with death and disease progression were calculated. RESULTS The analysis included a total of 2171 immunocompromised patients treated with RDV. The mean (standard deviation) age at index date was 75.1 (13.6) years. The median time to RDV initiation from hospitalization was 1.0 days (Q1-Q3: 1.0-2.0), and the median duration of RDV treatment was 5.0 days (Q1-Q3: 3.0-5.0). At RDV initiation, 53.02% (n=1151) of patients required non-invasive positive pressure ventilation, nasal high-flow or low-flow oxygen, and 2.26% (n=49) required mechanical ventilation, extracorporeal membrane oxygenation, or intensive care unit admission. Inpatient mortality rate by day 28 was 8.98% (95% confidence interval, 7.81-10.26). By day 28, the rate of disease progression was 9.86% (n=214), and 76.83% (n=1668) were discharged. CONCLUSION This study presents practical information on outcomes and treatment patterns of RDV in immunocompromised inpatients diagnosed with COVID-19.
Collapse
Affiliation(s)
- Manami Yoshida
- Gilead Sciences K. K, 1-9-2, Marunouchi, Chiyoda-ku, Tokyo, 100-6616, Japan.
| | - Nao Taguchi
- Gilead Sciences K. K, 1-9-2, Marunouchi, Chiyoda-ku, Tokyo, 100-6616, Japan
| | - Yi Piao
- Gilead Sciences K. K, 1-9-2, Marunouchi, Chiyoda-ku, Tokyo, 100-6616, Japan
| | - Rikisha Gupta
- Gilead Sciences Inc, 333, Lakeside Drive, Foster City, CA, 94404, USA
| | - Jami Peters
- Gilead Sciences Inc, 333, Lakeside Drive, Foster City, CA, 94404, USA
| | - Mazin Abdelghany
- Gilead Sciences Inc, 333, Lakeside Drive, Foster City, CA, 94404, USA
| | - Mel Chiang
- Gilead Sciences Inc, 333, Lakeside Drive, Foster City, CA, 94404, USA
| | - Chen-Yu Wang
- Gilead Sciences Inc, 333, Lakeside Drive, Foster City, CA, 94404, USA
| | - Mark Berry
- Gilead Sciences Inc, 333, Lakeside Drive, Foster City, CA, 94404, USA
| | - Hiroshi Yotsuyanagi
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of the Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| |
Collapse
|
8
|
Abt ER, Lam AK, Noguchi M, Rashid K, McLaughlin J, Teng PL, Tran W, Cheng D, Nesterenko PA, Mao Z, Creech AL, Burton Sojo G, Jeyachandran AV, Tam YK, Henley JE, Comai L, Pardi N, Arumugaswami V, Witte ON, Radu CG, Wu TT. Staggered immunization with mRNA vaccines encoding SARS-CoV-2 polymerase or spike antigens broadens the T cell epitope repertoire. Proc Natl Acad Sci U S A 2024; 121:e2406332121. [PMID: 39589869 PMCID: PMC11626164 DOI: 10.1073/pnas.2406332121] [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: 03/27/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Combining a T cell-targeting mRNA vaccine encoding the conserved SARS-CoV-2 RNA-dependent RNA polymerase, RdRp, with a Spike-encoding mRNA vaccine may offer an additional pathway toward COVID-19 protection. Here, we show that a nucleoside-modified RdRp mRNA vaccine raises robust and durable CD8+ T cell responses in mice. Immunization drives a CD8+ T cell response enriched toward a specific RdRp epitope. Unexpectedly, coadministration of mRNA vaccines encoding RdRp or the Spike Receptor Binding Domain (RBD) dampens RBD-specific immune responses. Contralateral administration reduces the suppression of RBD-specific T cell responses while type I interferon signaling blockade restores RBD-specific antibodies. A staggered immunization strategy maintains both RBD vaccine-mediated antibody and T cell responses as well as protection against lethal SARS-CoV-2 challenge in human ACE2 transgenic mice. In HLA-A2.1 transgenic mice, the RdRp vaccine elicits CD8+ T cell responses against HLA-A*02:01-restricted epitopes recognized by human donor T cells. These results highlight RdRp as a candidate antigen for COVID-19 vaccines. The findings also offer insights into crafting effective multivalent mRNA vaccines to broaden CD8+ T cell responses against SARS-CoV-2 and potentially other viruses with pandemic potential.
Collapse
Affiliation(s)
- Evan R. Abt
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Alex K. Lam
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Miyako Noguchi
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Khalid Rashid
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Jami McLaughlin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Pu-Lin Teng
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Wendy Tran
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Donghui Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Pavlo A. Nesterenko
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Zhiyuan Mao
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Amanda L. Creech
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Giselle Burton Sojo
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Arjit Vijey Jeyachandran
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Ying K. Tam
- Acuitas Therapeutics, Vancouver, BCV6T 1Z3, Canada
| | - Jill E. Henley
- Department of Molecular Microbiology and Immunology, The Hastings and Wright Laboratories, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
| | - Lucio Comai
- Department of Molecular Microbiology and Immunology, The Hastings and Wright Laboratories, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | | | - Owen N. Witte
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA90095
- Parker Institute for Cancer Immunotherapy, University of California Los Angeles, Los Angeles, CA90095
| | - Caius G. Radu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
| | - Ting-Ting Wu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
- AIDS Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA90095
| |
Collapse
|
9
|
Kocsmár É, Kocsmár I, Elamin F, Pápai L, Jakab Á, Várkonyi T, Glasz T, Rácz G, Pesti A, Danics K, Kiss A, Röst G, Belicza É, Schaff Z, Lotz G. Autopsy findings in cancer patients infected with SARS-CoV-2 show a milder presentation of COVID-19 compared to non-cancer patients. GeroScience 2024; 46:6101-6114. [PMID: 38691298 PMCID: PMC11493920 DOI: 10.1007/s11357-024-01163-7] [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: 12/18/2023] [Accepted: 04/12/2024] [Indexed: 05/03/2024] Open
Abstract
COVID-19, caused by SARS-CoV-2, manifests with differing severity across distinct patient subgroups, with outcomes influenced by underlying comorbidities such as cancer, which may cause functional and compositional alterations of the immune system during tumor progression. We aimed to investigate the association of SARS-CoV-2 infection and its complications with cancer in a large autopsy series and the role of COVID-19 in the fatal sequence leading to death. A total of 2641 adult autopsies were investigated, 539 of these were positive for SARS-CoV-2. Among the total number of patients analyzed, 829 had active cancer. Overall, the cohort included 100 patients who simultaneously had cancer and SARS-CoV-2 infection. The course of COVID-19 was less severe in cancer patients, including a significantly lower incidence of viral and bacterial pneumonia, occurring more frequently as a contributory disease or coexisting morbidity, or as SARS-CoV-2 positivity without viral disease. SARS-CoV-2 positivity was more frequent among non-metastatic than metastatic cancer cases, and in specific tumor types including hematologic malignancies. COVID-19 was more frequently found to be directly involved in the fatal sequence in patients undergoing active anticancer therapy, but less frequently in perioperative status, suggesting that the underlying malignancy and consequent surgery are more important factors leading to death perioperatively than viral disease. The course of COVID-19 in cancer patients was milder and balanced during the pandemic. This may be due to relative immunosuppressed status, and the fact that even early/mild viral infections can easily upset their condition, leading to death from their underlying cancer or its complications.
Collapse
Affiliation(s)
- Éva Kocsmár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary.
| | - Ildikó Kocsmár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
- Department of Urology, Semmelweis University, Budapest, Hungary
| | - Flóra Elamin
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Laura Pápai
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Ákos Jakab
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Tibor Várkonyi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Tibor Glasz
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Gergely Rácz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Adrián Pesti
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Krisztina Danics
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - András Kiss
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Gergely Röst
- National Laboratory for Health Security, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
| | - Éva Belicza
- Health Services Management Training Centre, Faculty of Health and Public Administration, Semmelweis University, Budapest, Hungary
| | - Zsuzsa Schaff
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Gábor Lotz
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
10
|
Carpp LN, Hyrien O, Fong Y, Benkeser D, Roels S, Stieh DJ, Van Dromme I, Van Roey GA, Kenny A, Huang Y, Carone M, McDermott AB, Houchens CR, Martins K, Jayashankar L, Castellino F, Amoa-Awua O, Basappa M, Flach B, Lin BC, Moore C, Naisan M, Naqvi M, Narpala S, O'Connell S, Mueller A, Serebryannyy L, Castro M, Wang J, Petropoulos CJ, Luedtke A, Lu Y, Yu C, Juraska M, Hejazi NS, Wolfe DN, Sadoff J, Gray GE, Grinsztejn B, Goepfert PA, Bekker LG, Gaur AH, Veloso VG, Randhawa AK, Andrasik MP, Hendriks J, Truyers C, Vandebosch A, Struyf F, Schuitemaker H, Douoguih M, Kublin JG, Corey L, Neuzil KM, Follmann D, Koup RA, Donis RO, Gilbert PB. Neutralizing antibody correlate of protection against severe-critical COVID-19 in the ENSEMBLE single-dose Ad26.COV2.S vaccine efficacy trial. Nat Commun 2024; 15:9785. [PMID: 39532861 PMCID: PMC11557889 DOI: 10.1038/s41467-024-53727-y] [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: 11/16/2023] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Assessment of immune correlates of severe COVID-19 has been hampered by the low numbers of severe cases in COVID-19 vaccine efficacy (VE) trials. We assess neutralizing and binding antibody levels at 4 weeks post-Ad26.COV2.S vaccination as correlates of risk and of protection against severe-critical COVID-19 through 220 days post-vaccination in the ENSEMBLE trial (NCT04505722), constituting ~4.5 months longer follow-up than our previous correlates analysis and enabling inclusion of 42 severe-critical vaccine-breakthrough cases. Neutralizing antibody titer is a strong inverse correlate of severe-critical COVID-19, with estimated hazard ratio (HR) per 10-fold increase 0.35 (95% CI: 0.13, 0.90). In a multivariable model, HRs are 0.31 (0.11, 0.89) for neutralizing antibody titer and 1.22 (0.49, 3.02) for anti-Spike binding antibody concentration. VE against severe-critical COVID-19 rises with neutralizing antibody titer: 63.1% (95% CI: 40.0%, 77.3%) at unquantifiable [<4.8975 International Units (IU)50/ml], 85.2% (47.2%, 95.3%) at just-quantifiable (5.2 IU50/ml), and 95.1% (81.1%, 96.9%) at 90th percentile (30.2 IU50/ml). At the same titers, VE against moderate COVID-19 is 32.5% (11.8%, 48.4%), 33.9% (19.1%, 59.3%), and 60.7% (40.4%, 76.4%). Protection against moderate vs. severe disease may require higher antibody levels, and very low antibody levels and/or other immune responses may associate with protection against severe disease.
Collapse
Affiliation(s)
- Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Sanne Roels
- Johnson & Johnson Innovative Medicine, Beerse, Belgium
| | - Daniel J Stieh
- Janssen Vaccines and Prevention, Leiden, the Netherlands
- Vaccine Company Inc., South San Francisco, CA, USA
| | | | | | - Avi Kenny
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
- Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Ying Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Marco Carone
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Sanofi Vaccines R&D, Marcy l'étoile, France
| | | | - Karen Martins
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | | | - Flora Castellino
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Obrimpong Amoa-Awua
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Manjula Basappa
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Britta Flach
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bob C Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Moore
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mursal Naisan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Muhammed Naqvi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah O'Connell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Allen Mueller
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Leo Serebryannyy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mike Castro
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Alex Luedtke
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Yiwen Lu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michal Juraska
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Nima S Hejazi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Daniel N Wolfe
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, the Netherlands
- Centivax, South San Francisco, CA, USA
| | - Glenda E Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Beatriz Grinsztejn
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Paul A Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Linda-Gail Bekker
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town, South Africa
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Observatory, Cape Town, South Africa
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Aditya H Gaur
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Valdilea G Veloso
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - April K Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michele P Andrasik
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jenny Hendriks
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Carla Truyers
- Johnson & Johnson Innovative Medicine, Beerse, Belgium
| | - An Vandebosch
- Johnson & Johnson Innovative Medicine, Beerse, Belgium
- argenx BV, Ghent, Belgium
| | - Frank Struyf
- Johnson & Johnson Innovative Medicine, Beerse, Belgium
- GSK, Wavre, Belgium
| | - Hanneke Schuitemaker
- Janssen Vaccines and Prevention, Leiden, the Netherlands
- Valneva, Saint-Herblain, France
| | - Macaya Douoguih
- Janssen Vaccines and Prevention, Leiden, the Netherlands
- Merck, Rahway, NJ, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
- Fogarty International Center, Bethesda, MD, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ruben O Donis
- Biomedical Advanced Research and Development Authority, Washington, DC, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA.
| |
Collapse
|
11
|
Lee JA, Han M, Ahn S, Lee Y, Yeom JS, Choi JY, Ku NS, Jeong SJ, Kim JH, Kim JS, Chung H, Cho H, Kim YR, Ahn JY. Long-Term Outcomes of COVID-19 and Risk Factors for Prolonged or Persistent COVID-19 in Lymphoma Patients: A Multicenter, Retrospective Cohort Study. J Korean Med Sci 2024; 39:e263. [PMID: 39468945 PMCID: PMC11519060 DOI: 10.3346/jkms.2024.39.e263] [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: 01/29/2024] [Accepted: 07/30/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Patients with hematologic malignancies exhibit persistent severe acute respiratory syndrome coronavirus 2 positivity over long periods after coronavirus disease 2019 (COVID-19) diagnosis. However, the frequency of, risk factors for, and prognosis of prolonged COVID-19 in immunocompromised patients remain unclear. Therefore, we investigated the long-term outcomes of COVID-19 in lymphoma patients and identified the associated factors and impact of prolonged COVID-19 on mortality. METHODS A multicenter retrospective cohort study of 583 lymphoma patients was conducted in 3 tertiary hospitals in South Korea. Patients receiving lymphoma treatment who were quarantined after obtaining a diagnosis of COVID-19 by polymerase chain reaction (PCR) or antigen test from August 2021 to September 2022 were examined. RESULTS Overall, 115 patients (19.7%) were diagnosed with COVID-19. Among 77 patients with clinical data, 24 had prolonged COVID-19. Patients in the prolonged COVID-19 group showed higher rates of receiving rituximab maintenance therapy following bendamustine and rituximab (BR) treatment for follicular lymphoma. This group did not show significant differences in clinical presentation within 30 days of COVID-19 diagnosis; however, it showed higher rates of re-admission due to COVID-19 pneumonia compared with the non-prolonged COVID-19 group. BR treatment followed by rituximab maintenance therapy is one of the risk factors for persistent PCR positivity, delayed or persistent pneumonia, and COVID-19 related admission after quarantine period. Prolonged COVID-19 was an independent risk factor for 1-year mortality. CONCLUSION Prolonged COVID-19 was more frequent in lymphoma patients who received BR treatment followed by rituximab maintenance therapy and associated with unfavorable long-term outcomes and higher 1-year mortality.
Collapse
Affiliation(s)
- Jung Ah Lee
- Division of Infectious Diseases, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Min Han
- Division of Infectious Diseases, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sangmin Ahn
- Division of Infectious Diseases, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yongseop Lee
- Division of Infectious Diseases, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Joon-Sup Yeom
- Division of Infectious Diseases, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jun Yong Choi
- Division of Infectious Diseases, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Nam Su Ku
- Division of Infectious Diseases, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Su Jin Jeong
- Division of Infectious Diseases, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Ho Kim
- Division of Infectious Diseases, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Seok Kim
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Haerim Chung
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyunsoo Cho
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yu Ri Kim
- Division of Hematology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| | - Jin Young Ahn
- Division of Infectious Diseases, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
12
|
Seng MSF, Ng KP, Soh TG, Tan TT, Chan M, Maiwald M, Tan LK, Linn YC, Leung W. A phase I/II study of adoptive SARS-CoV-2-specific T cells in immunocompromised hosts with or at risk of severe COVID-19 infection. Cytotherapy 2024; 26:1170-1178. [PMID: 38864802 DOI: 10.1016/j.jcyt.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Post-transplant or hematological cancer patients have a higher risk of mortality after infection with ancestral and early variants of severe acute respiratory syndrome (SARS)-CoV-2. Adoptive cell therapy (ACT) with virus-specific T cells (VSTs) could augment endogenous T cell immunity to avoid disease deterioration before viral clearance. METHODS We established a third-party SARS-CoV-2-specific T cell (COVID-T) bank in 2020 (NCT04351659) using convalescent and/or vaccinated donors. In a phase I/II study (NCT04457726), 13 adult and pediatric patients, acutely positive for SARS-CoV-2 and predicted to have a high chance of mortality, were recruited from September 2021 to February 2022. Twelve patients received a single dose of COVID-T cells, matched on at least 1 HLA. RESULTS A dose of either 75,000 or 150,000 IFN-γ+CD3+ cells/m2 SARS-COV-2-specific T cells did not cause cytokine release syndrome, acute respiratory distress syndrome, or graft-versus-host disease. In the 8 patients who had detectable donor SARS-COV-2-specific T cells after ACT, none progressed to severe disease or died with COVID-19. In contrast, among the other four patients without evidence of donor micro-chimerism, two died of COVID-19. CONCLUSIONS Long-acting third-party VSTs from convalescent or vaccinated donors could be expediently produced and might be clinically useful in future pandemics, particularly before global vaccination is implemented.
Collapse
Affiliation(s)
- Michaela Su-Fern Seng
- Department of Paediatric Hematology and Oncology, KK Women's and Children's Hospital, Singapore, Singapore; Duke-NUS Medical School, Singapore, Singapore
| | - King Pan Ng
- Department of Paediatric Hematology and Oncology, KK Women's and Children's Hospital, Singapore, Singapore; Duke-NUS Medical School, Singapore, Singapore
| | - Teck Guan Soh
- Department of Hematology, National University Hospital, Singapore, Singapore
| | - Thuan Tong Tan
- Duke-NUS Medical School, Singapore, Singapore; Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Marieta Chan
- HLA Laboratory, Health Sciences Authority, Singapore, Singapore
| | - Matthias Maiwald
- Duke-NUS Medical School, Singapore, Singapore; Department of Pathology and Laboratory Medicine, Microbiology Service, KK Women's and Children's Hospital, Singapore, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lip Kun Tan
- Department of Hematology, National University Hospital, Singapore, Singapore
| | - Yeh Ching Linn
- Duke-NUS Medical School, Singapore, Singapore; Department of Hematology, Singapore General Hospital, Singapore, Singapore
| | - Wing Leung
- Department of Paediatric Hematology and Oncology, KK Women's and Children's Hospital, Singapore, Singapore; Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
13
|
Notarbartolo S. T-Cell Immune Responses to SARS-CoV-2 Infection and Vaccination. Vaccines (Basel) 2024; 12:1126. [PMID: 39460293 PMCID: PMC11511197 DOI: 10.3390/vaccines12101126] [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: 09/04/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
The innate and adaptive immune systems collaborate to detect SARS-CoV-2 infection, minimize the viral spread, and kill infected cells, ultimately leading to the resolution of the infection. The adaptive immune system develops a memory of previous encounters with the virus, providing enhanced responses when rechallenged by the same pathogen. Such immunological memory is the basis of vaccine function. Here, we review the current knowledge on the immune response to SARS-CoV-2 infection and vaccination, focusing on the pivotal role of T cells in establishing protective immunity against the virus. After providing an overview of the immune response to SARS-CoV-2 infection, we describe the main features of SARS-CoV-2-specific CD4+ and CD8+ T cells, including cross-reactive T cells, generated in patients with different degrees of COVID-19 severity, and of Spike-specific CD4+ and CD8+ T cells induced by vaccines. Finally, we discuss T-cell responses to SARS-CoV-2 variants and hybrid immunity and conclude by highlighting possible strategies to improve the efficacy of COVID-19 vaccination.
Collapse
Affiliation(s)
- Samuele Notarbartolo
- Infectious Diseases Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
14
|
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
|
15
|
Rowntree LC, Audsley J, Allen LF, McQuilten HA, Hagen RR, Chaurasia P, Petersen J, Littler DR, Tan HX, Murdiyarso L, Habel JR, Foo IJH, Zhang W, Ten Berge ERV, Ganesh H, Kaewpreedee P, Lee KWK, Cheng SMS, Kwok JSY, Jayasinghe D, Gras S, Juno JA, Wheatley AK, Kent SJ, Rossjohn J, Cheng AC, Kotsimbos TC, Trubiano JA, Holmes NE, Pang Chan KK, Hui DSC, Peiris M, Poon LLM, Lewin SR, Doherty PC, Thevarajan I, Valkenburg SA, Kedzierska K, Nguyen THO. SARS-CoV-2-specific CD8 + T cells from people with long COVID establish and maintain effector phenotype and key TCR signatures over 2 years. Proc Natl Acad Sci U S A 2024; 121:e2411428121. [PMID: 39284068 PMCID: PMC11441481 DOI: 10.1073/pnas.2411428121] [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: 06/13/2024] [Accepted: 07/23/2024] [Indexed: 10/02/2024] Open
Abstract
Long COVID occurs in a small but important minority of patients following COVID-19, reducing quality of life and contributing to healthcare burden. Although research into underlying mechanisms is evolving, immunity is understudied. SARS-CoV-2-specific T cell responses are of key importance for viral clearance and COVID-19 recovery. However, in long COVID, the establishment and persistence of SARS-CoV-2-specific T cells are far from clear, especially beyond 12 mo postinfection and postvaccination. We defined ex vivo antigen-specific B cell and T cell responses and their T cell receptors (TCR) repertoires across 2 y postinfection in people with long COVID. Using 13 SARS-CoV-2 peptide-HLA tetramers, spanning 11 HLA allotypes, as well as spike and nucleocapsid probes, we tracked SARS-CoV-2-specific CD8+ and CD4+ T cells and B-cells in individuals from their first SARS-CoV-2 infection through primary vaccination over 24 mo. The frequencies of ORF1a- and nucleocapsid-specific T cells and B cells remained stable over 24 mo. Spike-specific CD8+ and CD4+ T cells and B cells were boosted by SARS-CoV-2 vaccination, indicating immunization, in fully recovered and people with long COVID, altered the immunodominance hierarchy of SARS-CoV-2 T cell epitopes. Meanwhile, influenza-specific CD8+ T cells were stable across 24 mo, suggesting no bystander-activation. Compared to total T cell populations, SARS-CoV-2-specific T cells were enriched for central memory phenotype, although the proportion of central memory T cells decreased following acute illness. Importantly, TCR repertoire composition was maintained throughout long COVID, including postvaccination, to 2 y postinfection. Overall, we defined ex vivo SARS-CoV-2-specific B cells and T cells to understand primary and recall responses, providing key insights into antigen-specific responses in people with long COVID.
Collapse
Affiliation(s)
- Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jennifer Audsley
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lilith F Allen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Ruth R Hagen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Priyanka Chaurasia
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Jan Petersen
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Dene R Littler
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lydia Murdiyarso
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jennifer R Habel
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Isabelle J H Foo
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Wuji Zhang
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Elizabeth R V Ten Berge
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hanujah Ganesh
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Prathanporn Kaewpreedee
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kelly W K Lee
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Samuel M S Cheng
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Janette S Y Kwok
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Dhilshan Jayasinghe
- Infection & Immunity Program, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3083, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3083, Australia
| | - Stephanie Gras
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Infection & Immunity Program, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3083, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3083, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Allen C Cheng
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
- Monash Infectious Diseases, Monash Health and School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Tom C Kotsimbos
- Department of Respiratory Medicine, The Alfred Hospital, Melbourne, VIC 3004, Australia
- Department of Medicine, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, VIC 3004, Australia
| | - Jason A Trubiano
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Department of Medicine (Austin Health), University of Melbourne, Heidelberg, VIC 3084, Australia
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia
| | - Natasha E Holmes
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia
- Department of Critical Care, University of Melbourne, Parkville, VIC 3000, Australia
- Data Analytics Research and Evaluation Centre, Austin Health and University of Melbourne, Heidelberg, VIC 3084, Australia
| | - Ken Ka Pang Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - David S C Hui
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Malik Peiris
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Leo L M Poon
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Sharon R Lewin
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Infectious Disease, Alfred Hospital and Monash University, Melbourne, VIC 3000, Australia
| | - Peter C Doherty
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Irani Thevarajan
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sophie A Valkenburg
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| |
Collapse
|
16
|
Martín-Escolano J, Salto-Alejandre S, Infante-Domínguez C, Carretero-Ledesma M, Maldonado-Lizarazo N, Camacho-Martínez P, Martín-Domínguez F, Tallón-Ruiz I, Ruiz-Molina A, Palacios-Baena Z, Pérez-Palacios P, Paniagua-García M, Álvarez-Marín R, Merino L, Cisneros JM, Cordero E, Pachón J, Pérez-Simón JA, Sánchez-Céspedes J, Aguilar-Guisado M. COVID-19 in patients with haematologic malignancies: Effect of RNAemia on clinical outcome in vaccinated patients. Int J Infect Dis 2024; 146:107163. [PMID: 38969329 DOI: 10.1016/j.ijid.2024.107163] [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: 03/04/2024] [Revised: 05/10/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024] Open
Abstract
OBJECTIVES Patients with haematologic malignancies (HM) COVID-19 have more severe disease, with increased risk of mortality. Therefore, this study aimed to evaluate the effect of SARS-CoV-2 RNAemia and the specific humoral immune responses on the clinical outcomes of patients with HM and COVID-19. METHODS Interferon-α/γ (IFN-α/IFN-γ) serum levels, neutralizing antibodies and RNAemia at COVID-19 diagnosis, and persistent RNAemia during the follow-up were evaluated. RESULTS Overall, 63 (58.9%) out of 107 patients had RNAemia, which was persistent in 26 (41.3%) patients. RNAemia at diagnosis and persistent RNAemia were associated with the need for high-flow nasal oxygen therapy during admission. Persistent RNAemia, age >70 years, and CURB-65 score ≥2 in patients with pneumonia were associated with increased 90-day mortality (P = 0.009, P = 0.030 and P = 0.001, respectively). The 90-day overall survival was lower (P = 0.006) in patients with persistent RNAemia. In addition, dexamethasone administration was associated with a COVID-19 episode with persistent RNAemia. CONCLUSION Our results suggest that in patients with HM, RNAemia at the time of COVID-19 diagnosis and during the follow-up can be used to stratify patients with HM according to their clinical evolution and to guide clinical decisions tailored to the specific needs of each patient.
Collapse
Affiliation(s)
- Javier Martín-Escolano
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Sonsoles Salto-Alejandre
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Carmen Infante-Domínguez
- Instituto de Biomedicina de Sevilla (IBiS), Hospitales Universitarios Virgen del Rocío y Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Carretero-Ledesma
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Natalia Maldonado-Lizarazo
- Instituto de Biomedicina de Sevilla (IBiS), Hospitales Universitarios Virgen del Rocío y Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain; Unidad de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Pedro Camacho-Martínez
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Francisco Martín-Domínguez
- Instituto de Biomedicina de Sevilla (IBiS), Hospitales Universitarios Virgen del Rocío y Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain; Servicio de Hematología, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | | | - Ana Ruiz-Molina
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Zaira Palacios-Baena
- Instituto de Biomedicina de Sevilla (IBiS), Hospitales Universitarios Virgen del Rocío y Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain; Unidad de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Patricia Pérez-Palacios
- Instituto de Biomedicina de Sevilla (IBiS), Hospitales Universitarios Virgen del Rocío y Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain; Unidad de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - María Paniagua-García
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Rocío Álvarez-Marín
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Merino
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - José Miguel Cisneros
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Elisa Cordero
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Jerónimo Pachón
- Instituto de Biomedicina de Sevilla (IBiS), Hospitales Universitarios Virgen del Rocío y Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - José Antonio Pérez-Simón
- Instituto de Biomedicina de Sevilla (IBiS), Hospitales Universitarios Virgen del Rocío y Virgen Macarena/CSIC/Universidad de Sevilla, Sevilla, Spain; Servicio de Hematología, Hospital Universitario Virgen del Rocío, Sevilla, Spain; Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Javier Sánchez-Céspedes
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain.
| | - Manuela Aguilar-Guisado
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
17
|
Spanjaart AM, Ljungman P, Tridello G, Schwartz J, Martinez-Cibrián N, Barba P, Kwon M, Lopez-Corral L, Martinez-Lopez J, Ferra C, Di Blasi R, Ghesquieres H, Mutsaers P, Calkoen F, Jak M, van Doesum J, Vermaat JSP, van der Poel M, Maertens J, Gambella M, Metafuni E, Ciceri F, Saccardi R, Nicholson E, Tholouli E, Matthew C, Potter V, Bloor A, Besley C, Roddie C, Wilson K, Nagler A, Campos A, Petersen SL, Folber F, Bader P, Finke J, Kroger N, Knelange N, de La Camara R, Kersten MJ, Mielke S. Improved outcome of COVID-19 over time in patients treated with CAR T-cell therapy: Update of the European COVID-19 multicenter study on behalf of the European Society for Blood and Marrow Transplantation (EBMT) Infectious Diseases Working Party (IDWP) and the European Hematology Association (EHA) Lymphoma Group. Leukemia 2024; 38:1985-1991. [PMID: 39043963 PMCID: PMC11347385 DOI: 10.1038/s41375-024-02336-1] [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: 04/03/2024] [Revised: 06/04/2024] [Accepted: 07/02/2024] [Indexed: 07/25/2024]
Abstract
COVID-19 has been associated with high mortality in patients treated with Chimeric Antigen Receptor (CAR) T-cell therapy for hematologic malignancies. Here, we investigated whether the outcome has improved over time with the primary objective of assessing COVID-19-attributable mortality in the Omicron period of 2022 compared to previous years. Data for this multicenter study were collected using the MED-A and COVID-19 report forms developed by the EBMT. One-hundred-eighty patients were included in the analysis, 39 diagnosed in 2020, 35 in 2021 and 106 in 2022. The median age was 58.9 years (min-max: 5.2-78.4). There was a successive decrease in COVID-19-related mortality over time (2020: 43.6%, 2021: 22.9%, 2022: 7.5%) and in multivariate analysis year of infection was the strongest predictor of survival (p = 0.0001). Comparing 2022 with 2020-2021, significantly fewer patients had lower respiratory symptoms (21.7% vs 37.8%, p = 0.01), needed oxygen support (25.5% vs 43.2%, p = 0.01), or were admitted to ICU (5.7% vs 33.8%, p = 0.0001). Although COVID-19-related mortality has decreased over time, CAR T-cell recipients remain at higher risk for complications than the general population. Consequently, vigilant monitoring for COVID-19 in patients undergoing B-cell-targeting CAR T-cell treatment is continuously recommended ensuring optimal prevention of infection and advanced state-of-the art treatment when needed.
Collapse
Affiliation(s)
- Anne Mea Spanjaart
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Cancer Center Amsterdam and LYMMCARE, Amsterdam, The Netherlands
| | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| | - Gloria Tridello
- European Society for Blood and Marrow Transplantation (EBMT) Data Office, Department of Medical Statistics & Bioinformatics, Leiden, Netherlands
| | - Juana Schwartz
- European Society for Blood and Marrow Transplantation (EBMT) Leiden Study Unit, European Society for Blood and Marrow Transplantation (EBMT) Data Office, Leiden, Netherlands
| | | | - Pere Barba
- Department of Hematology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Mi Kwon
- Department of Hematology, Hospital G. Universitario Gregorio Marañon, Institute of Health Research Gregorio Marañon, Madrid, Spain
| | - Lucia Lopez-Corral
- Department of Hematology, Hospital Universitario de Salamanca and IBSAL, Salamanca, Spain
| | - Joaquin Martinez-Lopez
- Department of Hematology, Hospital Univ. 12 de Octubre, Complutense University, CNIO, Madrid, Spain
| | - Christelle Ferra
- Clinical Hematology Department, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Barcelona, Spain
| | - Roberta Di Blasi
- Department of Hematology, Assistance Publique Hôpitaux de Paris-Hopital Saint-Louis, Paris, France
| | - Hervé Ghesquieres
- Department of Hematology, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Pim Mutsaers
- Department of Hematology, Erasmus MC Cancer Center, Rotterdam, the Netherlands
| | - Friso Calkoen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Margot Jak
- Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jaap van Doesum
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joost S P Vermaat
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Marjolein van der Poel
- Department of Hematology, Department of Internal Medicine, Division of Hematology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Johan Maertens
- Deptartment of Hematology, University Hospital Gasthuisberg, Leuven, Belgium
| | - Massimiliano Gambella
- Department of Hematology and Cellular Therapy, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Elisabetta Metafuni
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Fabio Ciceri
- Hematology and BMT Unit IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Riccardo Saccardi
- Cell Therapy and Transfusion Medicine Unit Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Emma Nicholson
- Department of Haematology, The Royal Marsden Hospital, London, United Kingdom
| | - Eleni Tholouli
- Department of Clinical Haematology, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Collin Matthew
- Adult HSCT unit, Northern Centre for Bone Marrow Transplantation, Newcastle Tyne, UK
| | - Victoria Potter
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, UK
| | - Adrian Bloor
- Adult Leukaemia and Bone Marrow Transplant Unit, Christie NHS Foundation Trust Hospital, University of Manchester, Manchester, UK
| | - Caroline Besley
- Department of Haematology, University Hospitals Bristol and Weston NHSFT, Bristol, UK
| | - Claire Roddie
- Department of Haematology, University College London Hospital, London, UK
| | - Keith Wilson
- Blood and Bone Marrow Transplantation Department, University Hospital of Cardiff, Cardiff, UK
| | - Arnon Nagler
- Chaim Sheba Medical Center, Tel Aviv University, Tel Hashomer, Israel
| | - Antonio Campos
- Celular Therapy Department, Instituto Portugués de Oncologia do Porto, Francisco Gentil, E.P.E, Porto, Portugal
| | - Soeren Lykke Petersen
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Frantisek Folber
- Department of internal Medicine, Hematology and Oncology, Masaryk University Hospital Brno, Brno, Czech Republic
| | - Peter Bader
- Department for Children and Adolescents, Division for Stem Cell Transplantation and Immunology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Jurgen Finke
- Department of Hematology/Oncology/Stem Cell Transplantation, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Nicolaus Kroger
- Department of Stem cell Transplantation, University Hospital Eppendorf, Hamburg, Germany
| | - Nina Knelange
- European Society for Blood and Marrow Transplantation (EBMT) Leiden Study Unit, European Society for Blood and Marrow Transplantation (EBMT) Data Office, Leiden, Netherlands
| | - Rafael de La Camara
- Department of Hematology, Hospital Universitario de La Princesa, Madrid, Spain
| | - Marie José Kersten
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Cancer Center Amsterdam and LYMMCARE, Amsterdam, The Netherlands
| | - Stephan Mielke
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Department of Laboratory Medicine, Karolinska Institutet and University Hospital, Karolinska Comprehensive Cancer Center, Karolinska ATMP Center, Stockholm, Sweden.
- Cellular Therapy and immunobiology working party (CTIWP) of EBMT, .
| |
Collapse
|
18
|
García Ramírez P, Callejas Charavia M, Oliva Martin R, Gómez La Hoz AM, Ortega MÁ, García Suárez J, Álvarez-Mon M, Monserrat Sanz J. SARS-CoV-2-Specific T Lymphocytes Analysis in mRNA-Vaccinated Patients with B-Cell Lymphoid Malignancies on Active Treatment. Vaccines (Basel) 2024; 12:961. [PMID: 39339993 PMCID: PMC11435597 DOI: 10.3390/vaccines12090961] [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: 07/02/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Patients with B-lymphocyte malignancies (BCMs) receiving B-lymphocyte-targeted therapies have increased risk of severe COVID-19 outcomes and impaired antibody response to SARS-CoV-2 mRNA vaccination in comparison to non-hematologic oncologic patients or general population. Consequently, it is vital to explore vaccine-induced T-lymphocyte responses in patients referred for the understanding of immune protection against SARS-CoV2 infections. The objective of the present study was to analyze the recall immune responses carried out by T lymphocytes after two COVID-19 mRNA vaccine doses. METHODS We enrolled 40 patients with BCMs and 10 healthy controls (HCs) after 4 weeks from the second mRNA vaccine dose. Spike (S)-specific T-lymphocyte responses were assessed in peripheral blood mononuclear lymphocytes (PBMCs) by intracellular IFN-γ staining combined with flow cytometry. Furthermore, the humoral response was assessed with the measurement of anti-spike antibodies. RESULTS From March to July 2021, 40 patients (median age 68) received mRNA vaccines. The overall antibody response for BCMs was 52.5% versus 100% for the healthy controls (p = 0.008). The antibody response was different across BCMs: 18.75% for non-Hodgkin lymphoma, 54.5% for chronic lymphocytic leukemia, and 92.3% for multiple myeloma. Responses varied by malignancy type and treatment, with anti-CD20 therapies showing the lowest response (6.7%). T-lymphocyte analysis revealed reduced numbers and altered differentiation stages in patients compared to the controls. However, the vaccine-induced T response was generally robust, with variations in specific T subpopulations. CONCLUSIONS mRNA vaccines induced significant humoral and cellular immune responses in B-cell lymphoid malignancy patients, although responses varied by treatment type and malignancy. Further research is needed to optimize vaccination strategies in this population.
Collapse
Affiliation(s)
- Patricia García Ramírez
- Hematology Department, University Hospital “Príncipe de Asturias”, Alcalá de Henares, 28805 Madrid, Spain;
| | - Marta Callejas Charavia
- Hematology Department, University Hospital “Príncipe de Asturias”, Alcalá de Henares, 28805 Madrid, Spain;
| | - Raquel Oliva Martin
- Department of Medicine, University of Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (R.O.M.); (A.M.G.L.H.); (M.Á.O.); (M.Á.-M.); (J.M.S.)
- IRYCIS Unit (Instituto Ramón y Cajal de Investigación Sanitaria), 28034 Madrid, Spain
| | - Ana María Gómez La Hoz
- Department of Medicine, University of Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (R.O.M.); (A.M.G.L.H.); (M.Á.O.); (M.Á.-M.); (J.M.S.)
- IRYCIS Unit (Instituto Ramón y Cajal de Investigación Sanitaria), 28034 Madrid, Spain
| | - Miguel Ángel Ortega
- Department of Medicine, University of Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (R.O.M.); (A.M.G.L.H.); (M.Á.O.); (M.Á.-M.); (J.M.S.)
- IRYCIS Unit (Instituto Ramón y Cajal de Investigación Sanitaria), 28034 Madrid, Spain
| | - Julio García Suárez
- Hematology Department, University Hospital “Príncipe de Asturias”, Alcalá de Henares, 28805 Madrid, Spain;
| | - Melchor Álvarez-Mon
- Department of Medicine, University of Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (R.O.M.); (A.M.G.L.H.); (M.Á.O.); (M.Á.-M.); (J.M.S.)
- IRYCIS Unit (Instituto Ramón y Cajal de Investigación Sanitaria), 28034 Madrid, Spain
| | - Jorge Monserrat Sanz
- Department of Medicine, University of Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (R.O.M.); (A.M.G.L.H.); (M.Á.O.); (M.Á.-M.); (J.M.S.)
- IRYCIS Unit (Instituto Ramón y Cajal de Investigación Sanitaria), 28034 Madrid, Spain
| |
Collapse
|
19
|
Leno-Duran E, Serrano-Conde E, Salas-Rodríguez A, Salcedo-Bellido I, Barrios-Rodríguez R, Fuentes A, Viñuela L, García F, Requena P. Evaluation of inflammatory biomarkers and their association with anti-SARS-CoV-2 antibody titers in healthcare workers vaccinated with BNT162B2. Front Immunol 2024; 15:1447317. [PMID: 39247198 PMCID: PMC11377239 DOI: 10.3389/fimmu.2024.1447317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/31/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction Vaccine-induced immunity against COVID-19 generates antibody and lymphocyte responses. However, variability in antibody titers has been observed after vaccination, and the determinants of a better response should be studied. The main objective of this investigation was to analyze the inflammatory biomarker response induced in healthcare workers vaccinated with BNT162b2, and its association with anti-Spike (a SARS-CoV-2 antigen) antibodies measured throughout a 1-year follow-up. Methods Anti-spike antibodies and 92 biomarkers were analyzed in serum, along with socio-demographic and clinical variables collected by interview or exploration. Results In our study, four biomarkers (ADA, IL-17C, CCL25 and CD8α) increased their expression after the first vaccine dose; and 8 others (uPA, IL-18R1, EN-RAGE, CASP-8, MCP-2, TNFβ, CD5 and CXCL10) decreased their expression. Age, body mass index (BMI), smoking, alcohol consumption, and prevalent diseases were associated with some of these biomarkers. Furthermore, higher baseline levels of T-cell surface glycoprotein CD6 and hepatocyte growth factor (HGF) were associated with lower mean antibody titers at follow-up, while levels of monocyte chemotactic protein 2 (MCP-2) had a positive association with antibody levels. Age and BMI were positively related to baseline levels of MCP-2 (β=0.02, 95%CI 0.00-0.04, p=0.036) and HGF (β=0.03, 95%CI 0.00-0.06, p=0.039), respectively. Conclusion Our findings indicate that primary BNT162b2 vaccination had a positive effect on the levels of several biomarkers related to T cell function, and a negative one on some others related to cancer or inflammatory processes. In addition, a higher level of MCP-2 and lower levels of HGF and CD6 were found to be associated with higher anti-Spike antibody titer following vaccination.
Collapse
Affiliation(s)
- Ester Leno-Duran
- Universidad de Granada, Departamento de Obstetricia y Ginecología, Granada, Spain
| | - Esther Serrano-Conde
- Servicio de Microbiología, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Ana Salas-Rodríguez
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain
| | - Inmaculada Salcedo-Bellido
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Rocío Barrios-Rodríguez
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Ana Fuentes
- Servicio de Microbiología, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Laura Viñuela
- Servicio de Microbiología, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Federico García
- Servicio de Microbiología, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Pilar Requena
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| |
Collapse
|
20
|
Hill JA, Martens MJ, Young JAH, Bhavsar K, Kou J, Chen M, Lee LW, Baluch A, Dhodapkar MV, Nakamura R, Peyton K, Howard DS, Ibrahim U, Shahid Z, Armistead P, Westervelt P, McCarty J, McGuirk J, Hamadani M, DeWolf S, Hosszu K, Sharon E, Spahn A, Toor AA, Waldvogel S, Greenberger LM, Auletta JJ, Horowitz MM, Riches ML, Perales MA. SARS-CoV-2 Vaccination in the First Year After Hematopoietic Cell Transplant or Chimeric Antigen Receptor T-Cell Therapy: A Prospective, Multicenter, Observational Study. Clin Infect Dis 2024; 79:542-554. [PMID: 38801746 PMCID: PMC11327798 DOI: 10.1093/cid/ciae291] [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: 05/03/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND The optimal timing of vaccination with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines after cellular therapy is incompletely understood. The objectives of this study are to determine whether humoral and cellular responses after SARS-CoV-2 vaccination differ if initiated <4 months versus 4-12 months after cellular therapy. METHODS We conducted a multicenter, prospective, observational study at 30 cancer centers in the United States. SARS-CoV-2 vaccination was administered as part of routine care. We obtained blood prior to and after vaccinations at up to 5 time points and tested for SARS-CoV-2 spike (anti-S) IgG in all participants and neutralizing antibodies for Wuhan D614G, Delta B.1.617.2, and Omicron B.1.1.529 strains, as well as SARS-CoV-2-specific T-cell receptors, in a subgroup. RESULTS We enrolled 466 allogeneic hematopoietic cell transplantation (HCT) (n = 231), autologous HCT (n = 170), and chimeric antigen receptor T-cell (CAR-T-cell) therapy (n = 65) recipients between April 2021 and June 2022. Humoral and cellular responses did not significantly differ among participants initiating vaccinations <4 months versus 4-12 months after cellular therapy. Anti-S IgG ≥2500 U/mL was correlated with high neutralizing antibody titers and attained by the last time point in 70%, 69%, and 34% of allogeneic HCT, autologous HCT, and CAR-T-cell recipients, respectively. SARS-CoV-2-specific T-cell responses were attained in 57%, 83%, and 58%, respectively. Pre-cellular therapy SARS-CoV-2 infection or vaccination and baseline B-cell count were key predictors of post-cellular therapy immunity. CONCLUSIONS These data support mRNA SARS-CoV-2 vaccination prior to, and reinitiation 3 to 4 months after, cellular therapies with allogeneic HCT, autologous HCT, and CAR-T-cell therapy.
Collapse
Grants
- Kyowa Kirin
- OptumHealth
- Takeda Oncology Co
- Xenikos BV
- Talaris Therapeutics
- Karyopharm Therapeutics
- Sanofi Genzyme
- MorphoSys
- U10HL069294 National Cancer Institute [NCI]
- Karius
- Vertex
- OncoImmune, Inc
- Orca Biosystems, Inc
- Medexus, Merck & Co.
- Kyowa Kirin International plc
- Seagen, Inc
- P30 CA015704 NCI NIH HHS
- Bristol Myers Squibb Co
- HistoGenetics
- Millennium
- U24 CA076518 NCI NIH HHS
- Oncopeptides, Inc
- Janssen Research & Development, LLC
- Miltenyi Biotec, Inc
- AlloVir, Inc
- UG1 HL069315 NHLBI NIH HHS
- Janssen/Johnson & Johnson
- UG1 HL138645 NHLBI NIH HHS
- HRSA HHS
- Terumo Blood and Cell Technologies
- N00014-20-1-2705 Department of Health and Human Services [DHHS]
- Adienne SA
- Novartis
- Kiadis Pharma
- Actinium Pharmaceuticals, Inc
- Bluebird Bio, Inc
- Novartis Pharmaceuticals Corporation
- Medical College of Wisconsin
- HHSH234200637015C National Institute of Allergy and Infectious Diseases
- UG1 HL069246 NHLBI NIH HHS
- TG Therapeutics
- Pfizer, Inc
- Kite Pharma, Inc
- P30 CA008748 NCI NIH HHS
- Incyte Corporation
- Pharmacyclics, LLC
- Tscan
- National Heart, Lung, and Blood Institute [NHLBI]
- CytoSen Therapeutics, Inc
- Gilead
- Astellas Pharma US
- Takeda Pharmaceuticals
- Accenture
- AbbVie
- Gilead Company
- Be the Match Foundation
- Leukemia and Lymphoma Society
- Adaptive Biotechnologies
- National Marrow Donor Program/Be the Match
- Multiple Myeloma Research Foundation
- Stemcyte
- DBA Eurofins Transplant Diagnostics
- CareDx
- Eurofins Viracor
- NIH HHS
- CSL Behring
- Medac GmbH
- GlaxoSmithKline
- Fate Therapeutics
- American Society for Transplantation and Cellular Therapy
- Gamida-Cell, Ltd
- NCI
- Legend Biotech
- Kadmon
- Ossium Health, Inc
- Vor Biopharma
- Jasper Therapeutics
- Jazz Pharmaceuticals, Inc
- Iovance
- U24CA076518
- LabCorp
- Omeros Corporation
- Amgen, Inc
- Magenta Therapeutics
- Daiichi Sankyo Co, Ltd
- Priothera
- Office of Naval Research
- National Institutes of Health
- Health Resources and Services Administration
- Janssen Research & Development, LLC
- Janssen/Johnson & Johnson
- Medexus, Merck & Co.
Collapse
Affiliation(s)
- Joshua A Hill
- Vaccine and Infectious Disease, Fred Hutchinson Cancer Center, and Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Michael J Martens
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jo-Anne H Young
- Division of Infectious Diseases, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kavita Bhavsar
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jianqun Kou
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Min Chen
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Lik Wee Lee
- Adaptive Biotechnologies Corporation, Seattle, Washington, USA
| | - Aliyah Baluch
- Division of Infectious Diseases, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Ryotaro Nakamura
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, Duarte, California, USA
| | | | - Dianna S Howard
- Division of Hematology and Oncology, Wake Forest Baptist, Winston-Salem, North Carolina, USA
| | - Uroosa Ibrahim
- Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, New York, USA
| | - Zainab Shahid
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Paul Armistead
- Division of Hematology, University of North Carolina Medical Center, Chapel Hill, North Carolina, USA
| | - Peter Westervelt
- Division of Oncology, Barnes-Jewish Hospital, Washington University, St. Louis, Missouri, USA
| | - John McCarty
- Division of Hematology, Oncology & Palliative Care, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Joseph McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas, Lawrence, Kansas, USA
| | - Mehdi Hamadani
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Susan DeWolf
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kinga Hosszu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elad Sharon
- National Cancer Institute, Bethesda, Maryland, USA
| | - Ashley Spahn
- National Marrow Donor Program/Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota, USA
| | - Amir A Toor
- Lehigh Valley Health Network, Allentown, Pennsylvania, USA
| | - Stephanie Waldvogel
- National Marrow Donor Program/Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota, USA
| | - Lee M Greenberger
- The Leukemia and Lymphoma Society, Rye Brook, New York, New York, USA
| | - Jeffery J Auletta
- National Marrow Donor Program/Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota, USA
- Division of Hematology/Oncology/BMT and Infectious Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Mary M Horowitz
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Marcie L Riches
- Center for International Blood and Marrow Transplantation Research, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Miguel-Angel Perales
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
21
|
Grosso D, Wagner JL, O’Connor A, Keck K, Huang Y, Wang ZX, Mehler H, Leiby B, Flomenberg P, Gergis U, Nikbakht N, Morris M, Karp J, Peedin A, Flomenberg N. Safety and feasibility of third-party cytotoxic T lymphocytes for high-risk patients with COVID-19. Blood Adv 2024; 8:4113-4124. [PMID: 38885482 PMCID: PMC11345373 DOI: 10.1182/bloodadvances.2024013344] [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: 04/08/2024] [Revised: 05/24/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
ABSTRACT Cytotoxic T lymphocytes (CTLs) destroy virally infected cells and are critical for the elimination of viral infections such as those caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Delayed and dysfunctional adaptive immune responses to SARS-CoV-2 are associated with poor outcomes. Treatment with allogeneic SARS-CoV-2-specific CTLs may enhance cellular immunity in high-risk patients providing a safe, direct mechanism of treatment. Thirty high-risk ambulatory patients with COVID-19 were enrolled in a phase 1 trial assessing the safety of third party, SARS-CoV-2-specific CTLs. Twelve interventional patients, 6 of whom were immunocompromised, matched the HLA-A∗02:01 restriction of the CTLs and received a single infusion of 1 of 4 escalating doses of a product containing 68.5% SARS-CoV-2-specific CD8+ CTLs/total cells. Symptom improvement and resolution in these patients was compared with an observational group of 18 patients lacking HLA-A∗02:01 who could receive standard of care. No dose-limiting toxicities were observed at any dosing level. Nasal swab polymerase chain reaction testing showed ≥88% and >99% viral elimination from baseline in all patients at 4 and 14 days after infusion, respectively. The CTLs did not interfere with the development of endogenous anti-SARS-CoV-2 humoral or cellular responses. T-cell receptor β analysis showed persistence of donor-derived SARS-CoV-2-specific CTLs through the end of the 6-month follow-up period. Interventional patients consistently reported symptomatic improvement 2 to 3 days after infusion, whereas improvement was more variable in observational patients. SARS-CoV-2-specific CTLs are a potentially feasible cellular therapy for COVID-19 illness. This trial was registered at www.clinicaltrials.gov as #NCT04765449.
Collapse
Affiliation(s)
- Dolores Grosso
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - John L. Wagner
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Allyson O’Connor
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Kaitlyn Keck
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Yanping Huang
- Department of Pathology and Genomic Medicine, Histocompatibility and Immunogenetics Laboratory, Thomas Jefferson University, Philadelphia, PA
| | - Zi-Xuan Wang
- Departments of Surgery and Pathology, Molecular and Genomic Pathology Laboratory, Thomas Jefferson University, Philadelphia, PA
| | - Hilary Mehler
- Department of Pathology and Genomic Medicine, Histocompatibility and Immunogenetics Laboratory, Thomas Jefferson University, Philadelphia, PA
| | - Benjamin Leiby
- Division of Biostatistics, Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Phyllis Flomenberg
- Division of Infectious Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Usama Gergis
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Neda Nikbakht
- Department of Dermatology and Cutaneous Biology, Cutaneous Lymphoma Clinic, Thomas Jefferson University, Philadelphia, PA
| | - Michael Morris
- Department of Emergency Medicine, Thomas Jefferson University Washington Township Hospital, Sewell, NJ
| | - Julie Karp
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Alexis Peedin
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Neal Flomenberg
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
22
|
Martín-Sánchez E, Tamariz-Amador LE, Guerrero C, Zherniakova A, Zabaleta A, Maia C, Blanco L, Alignani D, Fortuño MA, Grande C, Manubens A, Arguiñano JM, Gomez C, Perez-Persona E, Olazabal I, Oiartzabal I, Panizo C, Prosper F, San-Miguel JF, Rodriguez-Otero P, Paiva B. Immune dysfunction prior to and during vaccination in multiple myeloma: a case study based on COVID-19. Blood Cancer J 2024; 14:111. [PMID: 38987557 PMCID: PMC11237013 DOI: 10.1038/s41408-024-01089-5] [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: 03/26/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/12/2024] Open
Abstract
Infection is the leading cause of death in multiple myeloma (MM). However, the cellular composition associated with immune dysfunction is not defined. We analyzed immune profiles in the peripheral blood of patients with MM (n = 28) and B-cell chronic lymphoproliferative disorders (n = 53) vs. health care practitioners (n = 96), using multidimensional and computational flow cytometry. MM patients displayed altered distribution of most cell types (41/56, 73%), particularly within the B-cell (17/17) and T-cell (20/30) compartments. Using COVID-19 as a case study, we compared the immune response to vaccination based on 64,304 data points generated from the analysis of 1099 longitudinal samples. MM patients showed limited B-cell expansion linked to lower anti-RBD and anti-S antibody titers after the first two doses and booster. The percentages of B cells and CD4+ T cells in the blood, as well as the absolute counts of B cells and dendritic cells, predicted vaccine immunogenicity at different time points. In contrast with the humoral response, the percentage and antigen-dependent differentiation of SARS-CoV-2-specific CD8+ T cells was not altered in MM patients. Taken together, this study defined the cellular composition associated with immune dysfunction in MM and provided biomarkers such as the B-cell percentage and absolute count to individualize vaccination calendars.
Collapse
Affiliation(s)
- Esperanza Martín-Sánchez
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain.
| | - Luis-Esteban Tamariz-Amador
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Camila Guerrero
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Anastasiia Zherniakova
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Aintzane Zabaleta
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Catarina Maia
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Laura Blanco
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Diego Alignani
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Maria-Antonia Fortuño
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Carlos Grande
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Andrea Manubens
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | | | - Clara Gomez
- Hospital Universitario de Galdakao, Galdakano, Spain
| | | | - Iñigo Olazabal
- Hospital Universitario de Donostia, San Sebastian, Spain
| | | | - Carlos Panizo
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
- Hospital Universitario de Donostia, San Sebastian, Spain
| | - Felipe Prosper
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Jesus F San-Miguel
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Paula Rodriguez-Otero
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Bruno Paiva
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IdiSNA), CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain.
| |
Collapse
|
23
|
Mülling N, Behr FM, Heieis GA, Boss K, van Duikeren S, van Haften FJ, Pardieck IN, van der Gracht ET, Vleeshouwers W, van der Sluis TC, de Graaf JF, Veerkamp DM, Franken KL, Lei X, van de Sand L, van der Burg SH, Welters MJ, Heidt S, Huisman W, Jochems SP, Giera M, Witzke O, de Vries AP, Kribben A, Everts B, Wilde B, Arens R. Inhibiting the NADase CD38 improves cytomegalovirus-specific CD8+ T cell functionality and metabolism. J Clin Invest 2024; 134:e179561. [PMID: 38954588 PMCID: PMC11364383 DOI: 10.1172/jci179561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
Cytomegalovirus (CMV) is one of the most common and relevant opportunistic pathogens in people who are immunocompromised, such as kidney transplant recipients (KTRs). The exact mechanisms underlying the disability of cytotoxic T cells to provide sufficient protection against CMV in people who are immunosuppressed have not been identified yet. Here, we performed in-depth metabolic profiling of CMV-specific CD8+ T cells in patients who are immunocompromised and show the development of metabolic dysregulation at the transcriptional, protein, and functional level of CMV-specific CD8+ T cells in KTRs with noncontrolled CMV infection. These dysregulations comprise impaired glycolysis and increased mitochondrial stress, which is associated with an intensified expression of the nicotinamide adenine dinucleotide nucleotidase (NADase) CD38. Inhibiting NADase activity of CD38 reinvigorated the metabolism and improved cytokine production of CMV-specific CD8+ T cells. These findings were corroborated in a mouse model of CMV infection under conditions of immunosuppression. Thus, dysregulated metabolic states of CD8+ T cells could be targeted by inhibiting CD38 to reverse hyporesponsiveness in individuals who fail to control chronic viral infection.
Collapse
Affiliation(s)
- Nils Mülling
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Felix M. Behr
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Graham A. Heieis
- Department of Parasitology, Leiden University Centre for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Kristina Boss
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Suzanne van Duikeren
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Iris N. Pardieck
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ward Vleeshouwers
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | - Kees L.M.C. Franken
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Xin Lei
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lukas van de Sand
- Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | | | | | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wesley Huisman
- Department of Parasitology, Leiden University Centre for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Simon P. Jochems
- Department of Parasitology, Leiden University Centre for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Oliver Witzke
- Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Aiko P.J. de Vries
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Andreas Kribben
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Bart Everts
- Department of Parasitology, Leiden University Centre for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Benjamin Wilde
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ramon Arens
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
24
|
Fan X, Song JW, Cao WJ, Zhou MJ, Yang T, Wang J, Meng FP, Shi M, Zhang C, Wang FS. T-Cell Epitope Mapping of SARS-CoV-2 Reveals Coordinated IFN-γ Production and Clonal Expansion of T Cells Facilitates Recovery from COVID-19. Viruses 2024; 16:1006. [PMID: 39066169 PMCID: PMC11281491 DOI: 10.3390/v16071006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/01/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND T-cell responses can be protective or detrimental during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection; however, the underlying mechanism is poorly understood. METHODS In this study, we screened 144 15-mer peptides spanning the SARS-CoV-2 spike, nucleocapsid (NP), M, ORF8, ORF10, and ORF3a proteins and 39 reported SARS-CoV-1 peptides in peripheral blood mononuclear cells (PBMCs) from nine laboratory-confirmed coronavirus disease 2019 (COVID-19) patients (five moderate and four severe cases) and nine healthy donors (HDs) collected before the COVID-19 pandemic. T-cell responses were monitored by IFN-γ and IL-17A production using ELISA, and the positive samples were sequenced for the T cell receptor (TCR) β chain. The positive T-cell responses to individual SARS-CoV-2 peptides were validated by flow cytometry. RESULTS COVID-19 patients with moderate disease produced more IFN-γ than HDs and patients with severe disease (moderate vs. HDs, p < 0.0001; moderate vs. severe, p < 0.0001) but less IL-17A than those with severe disease (p < 0.0001). A positive correlation was observed between IFN-γ production and T-cell clonal expansion in patients with moderate COVID-19 (r = 0.3370, p = 0.0214) but not in those with severe COVID-19 (r = -0.1700, p = 0.2480). Using flow cytometry, we identified that a conserved peptide of the M protein (Peptide-120, P120) was a dominant epitope recognized by CD8+ T cells in patients with moderate disease. CONCLUSION Coordinated IFN-γ production and clonal expansion of SARS-CoV-2-specific T cells are associated with disease resolution in COVID-19. Our findings contribute to a better understanding of T-cell-mediated immunity in COVID-19 and may inform future strategies for managing and preventing severe outcomes of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xing Fan
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Jin-Wen Song
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
- Medical School of Chinese PLA, Beijing 100853, China
| | - Wen-Jing Cao
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ming-Ju Zhou
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Tao Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Jing Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Fan-Ping Meng
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Ming Shi
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Chao Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
- Medical School of Chinese PLA, Beijing 100853, China
| |
Collapse
|
25
|
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
|
26
|
Ng HJ, Alata MK, Nguyen QT, Huynh Duc Vinh P, Tan JY, Wong CL. Managing and treating COVID-19 in patients with hematological malignancies: a narrative review and expert insights. Clin Exp Med 2024; 24:119. [PMID: 38833206 PMCID: PMC11150206 DOI: 10.1007/s10238-024-01381-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/18/2024] [Indexed: 06/06/2024]
Abstract
Patients with hematologic malignancies (HMs) are at a significantly higher risk of contracting COVID-19 and experiencing severe outcomes compared to individuals without HMs. This heightened risk is influenced by various factors, including the underlying malignancy, immunosuppressive treatments, and patient-related factors. Notably, immunosuppressive regimens commonly used for HM treatment can lead to the depletion of B cells and T cells, which is associated with increased COVID-19-related complications and mortality in these patients. As the pandemic transitions into an endemic state, it remains crucial to acknowledge and address the ongoing risk for individuals with HMs. In this review, we aim to summarize the current evidence to enhance our understanding of the impact of HMs on COVID-19 risks and outcomes, identify particularly vulnerable individuals, and emphasize the need for specialized clinical attention and management. Furthermore, the impaired immune response to COVID-19 vaccination observed in these patients underscores the importance of implementing additional mitigation strategies. This may include targeted prophylaxis and treatment with antivirals and monoclonal antibodies as indicated. To provide practical guidance and considerations, we present two illustrative cases to highlight the real-life challenges faced by physicians caring for patients with HMs, emphasizing the need for individualized management based on disease severity, type, and the unique circumstances of each patient.
Collapse
Affiliation(s)
- Heng Joo Ng
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | | | - Quang The Nguyen
- Stem Cell Transplantation Department, Blood Transfusion Hematology Hospital, Ho Chi Minh, Vietnam
| | - Phu Huynh Duc Vinh
- Stem Cell Transplantation Department, Blood Transfusion Hematology Hospital, Ho Chi Minh, Vietnam
| | - Jing Yuan Tan
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Chieh Lee Wong
- Department of Haematology, Sunway Medical Centre, Bandar Sunway, Selangor, Malaysia.
- School of Medical and Life Sciences, Sunway University, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
27
|
Singer J, Tunbridge MJ, Shi B, Perkins GB, Chai CS, Salehi T, Sim BZ, Kireta S, Johnston JK, Akerman A, Milogiannakis V, Aggarwal A, Turville S, Hissaria P, Ying T, Wu H, Grubor-Bauk B, Coates PT, Chadban SJ. Dietary Inulin to Improve SARS-CoV-2 Vaccine Response in Kidney Transplant Recipients: The RIVASTIM-Inulin Randomised Controlled Trial. Vaccines (Basel) 2024; 12:608. [PMID: 38932337 PMCID: PMC11209582 DOI: 10.3390/vaccines12060608] [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: 05/02/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Kidney transplant recipients are at an increased risk of hospitalisation and death from SARS-CoV-2 infection, and standard two-dose vaccination schedules are typically inadequate to generate protective immunity. Gut dysbiosis, which is common among kidney transplant recipients and known to effect systemic immunity, may be a contributing factor to a lack of vaccine immunogenicity in this at-risk cohort. The gut microbiota modulates vaccine responses, with the production of immunomodulatory short-chain fatty acids by bacteria such as Bifidobacterium associated with heightened vaccine responses in both observational and experimental studies. As SCFA-producing populations in the gut microbiota are enhanced by diets rich in non-digestible fibre, dietary supplementation with prebiotic fibre emerges as a potential adjuvant strategy to correct dysbiosis and improve vaccine-induced immunity. In a randomised, double-bind, placebo-controlled trial of 72 kidney transplant recipients, we found dietary supplementation with prebiotic inulin for 4 weeks before and after a third SARS-CoV2 mRNA vaccine to be feasible, tolerable, and safe. Inulin supplementation resulted in an increase in gut Bifidobacterium, as determined by 16S RNA sequencing, but did not increase in vitro neutralisation of live SARS-CoV-2 virus at 4 weeks following a third vaccination. Dietary fibre supplementation is a feasible strategy with the potential to enhance vaccine-induced immunity and warrants further investigation.
Collapse
Affiliation(s)
- Julian Singer
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia; (J.S.); (T.Y.); (H.W.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Matthew J. Tunbridge
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
| | - Bree Shi
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Griffith B. Perkins
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; (G.B.P.); (C.S.C.); (P.H.); (B.G.-B.)
- Immunology Directorate, SA Pathology, Adelaide, SA 5000, Australia
| | - Cheng Sheng Chai
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; (G.B.P.); (C.S.C.); (P.H.); (B.G.-B.)
| | - Tania Salehi
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
| | - Beatrice Z. Sim
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
| | - Svjetlana Kireta
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
| | - Julie K. Johnston
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
| | - Anouschka Akerman
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (A.A.); (V.M.); (A.A.); (S.T.)
| | - Vanessa Milogiannakis
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (A.A.); (V.M.); (A.A.); (S.T.)
| | - Anupriya Aggarwal
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (A.A.); (V.M.); (A.A.); (S.T.)
| | - Stuart Turville
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (A.A.); (V.M.); (A.A.); (S.T.)
| | - Pravin Hissaria
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; (G.B.P.); (C.S.C.); (P.H.); (B.G.-B.)
- Department of Immunology and Allergy, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Tracey Ying
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia; (J.S.); (T.Y.); (H.W.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Huiling Wu
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia; (J.S.); (T.Y.); (H.W.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| | - Branka Grubor-Bauk
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; (G.B.P.); (C.S.C.); (P.H.); (B.G.-B.)
- Viral Immunology Group, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Adelaide, SA 5011, Australia
| | - P. Toby Coates
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; (M.J.T.); (T.S.); (B.Z.S.); (S.K.); (J.K.J.); (P.T.C.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia; (G.B.P.); (C.S.C.); (P.H.); (B.G.-B.)
| | - Steven J. Chadban
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia; (J.S.); (T.Y.); (H.W.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia;
| |
Collapse
|
28
|
Glueck OM, Liang X, Badell I, Wratil PR, Graf A, Krebs S, Blum H, Hellmuth JC, Scherer C, Hollaus A, Spaeth PM, Karakoc B, Fuchs T, Zimmermann J, Kauke T, Moosmann A, Keppler OT, Schneider C, Muenchhoff M. Impaired immune responses and prolonged viral replication in lung allograft recipients infected with SARS-CoV-2 in the early phase after transplantation. Infection 2024; 52:847-855. [PMID: 37922037 PMCID: PMC11143031 DOI: 10.1007/s15010-023-02116-6] [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: 07/26/2023] [Accepted: 10/12/2023] [Indexed: 11/05/2023]
Abstract
PURPOSE Lung transplant recipients are at increased risk of severe disease following infection with severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) due to high-dose immunosuppressive drugs and the lung is the main organ affected by Coronavirus disease 2019 (COVID-19). Several studies have confirmed increased SARS-CoV-2-related mortality and morbidity in patients living with lung allografts; however, detailed immunological studies of patients with SARS-CoV-2 infection in the early phase following transplantation remain scarce. METHODS We investigated patients who were infected with SARS-CoV-2 in the early phase (18-103 days) after receiving double-lung allografts (n = 4, LuTx) in comparison to immunocompetent patients who had not received solid organ transplants (n = 88, noTx). We analyzed SARS-CoV-2-specific antibody responses against the SARS-CoV-2 spike and nucleocapsid proteins using enzyme-linked immunosorbent assays (ELISA), chemiluminescence immunoassays (CLIA), and immunoblot assays. T cell responses were investigated using Elispot assays. RESULTS One LuTx patient suffered from persistent infection with fatal outcome 122 days post-infection despite multiple interventions including remdesivir, convalescent plasma, and the monoclonal antibody bamlanivimab. Two patients experienced clinically mild disease with prolonged viral shedding (47 and 79 days), and one patient remained asymptomatic. Antibody and T cell responses were significantly reduced or undetectable in all LuTx patients compared to noTx patients. CONCLUSION Patients in the early phase following lung allograft transplantation are vulnerable to infection with SARS-CoV-2 due to impaired immune responses. This patient population should be vaccinated before LuTx, protected from infection post-LuTx, and in case of infection treated generously with currently available interventions.
Collapse
Affiliation(s)
- Olaf M Glueck
- Division of Thoracic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Xiaoling Liang
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig Maximilian University of Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
| | - Irina Badell
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig Maximilian University of Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
| | - Paul R Wratil
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig Maximilian University of Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Alexander Graf
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilian University of Munich, Munich, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilian University of Munich, Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, Ludwig Maximilian University of Munich, Munich, Germany
| | - Johannes C Hellmuth
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Clemens Scherer
- Department of Medicine I, LMU University Hospital, LMU Munich, Munich, Germany
| | - Alexandra Hollaus
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Helmholtz Munich, Munich, Germany
| | - Patricia M Spaeth
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig Maximilian University of Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
| | - Burak Karakoc
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig Maximilian University of Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
| | - Thimo Fuchs
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig Maximilian University of Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
| | - Julia Zimmermann
- Division of Thoracic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Teresa Kauke
- Division of Thoracic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Andreas Moosmann
- German Center for Infection Research (DZIF), Partner Site, Munich, Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Helmholtz Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Oliver T Keppler
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig Maximilian University of Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site, Munich, Germany
| | - Christian Schneider
- Division of Thoracic Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Maximilian Muenchhoff
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig Maximilian University of Munich, Pettenkoferstr. 9a, 80336, Munich, Germany.
- German Center for Infection Research (DZIF), Partner Site, Munich, Germany.
| |
Collapse
|
29
|
Pourriyahi H, Hajizadeh N, Khosravi M, Pourriahi H, Soleimani S, Hosseini NS, Mohammad AP, Goodarzi A. New variants of COVID-19 (XBB.1.5 and XBB.1.16, the "Arcturus"): A review of highly questioned concerns, a brief comparison between different peaks in the COVID-19 pandemic, with a focused systematic review on expert recommendations for prevention, vaccination, and treatment measures in the general population and at-risk groups. Immun Inflamm Dis 2024; 12:e1323. [PMID: 38938013 PMCID: PMC11211615 DOI: 10.1002/iid3.1323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/11/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024] Open
Abstract
INTRODUCTION The COVID-19 pandemic has taken many forms and continues to evolve, now around the Omicron wave, raising concerns over the globe. With COVID-19 being declared no longer a "public health emergency of international concern (PHEIC)," the COVID pandemic is still far from over, as new Omicron subvariants of interest and concern have risen since January of 2023. Mainly with the XBB.1.5 and XBB.1.16 subvariants, the pandemic is still very much "alive" and "breathing." METHODS This review consists of five highly concerning questions about the current state of the COVID Omicron peak. We searched four main online databases to answer the first four questions. For the last one, we performed a systematic review of the literature, with keywords "Omicron," "Guidelines," and "Recommendations." RESULTS A total of 31 articles were included. The main symptoms of the current Omicron wave include a characteristically high fever, coughing, conjunctivitis (with itching eyes), sore throat, runny nose, congestion, fatigue, body ache, and headache. The median incubation period of the symptoms is shorter than the previous peaks. Vaccination against COVID can still be considered effective for the new subvariants. CONCLUSION Guidelines recommend continuation of personal protective measures, third and fourth dose boosters, along with administration of bivalent messenger RNA vaccine boosters. The consensus antiviral treatment is combination therapy using Nirmatrelvir and Ritonavir, and the consensus for pre-exposure prophylaxis is Tixagevimab and Cilgavimab combination. We hope the present paper raises awareness for the continuing presence of COVID and ways to lower the risks, especially for at-risk groups.
Collapse
Affiliation(s)
- Homa Pourriyahi
- Student Research Committee, School of MedicineIran University of Medical SciencesTehranIran
| | - Nima Hajizadeh
- School of MedicineIran University of Medical SciencesTehranIran
| | - Mina Khosravi
- School of MedicineIran University of Medical SciencesTehranIran
| | - Homayoun Pourriahi
- Student Research Committee, School of MedicineIran University of Medical SciencesTehranIran
| | - Sanaz Soleimani
- Student Research Committee, School of MedicineIran University of Medical SciencesTehranIran
- Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of MedicineIran University of Medical SciencesTehranIran
| | | | | | - Azadeh Goodarzi
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of MedicineIran University of Medical SciencesTehranIran
| |
Collapse
|
30
|
Delmonte OM, Oguz C, Dobbs K, Myint-Hpu K, Palterer B, Abers MS, Draper D, Truong M, Kaplan IM, Gittelman RM, Zhang Y, Rosen LB, Snow AL, Dalgard CL, Burbelo PD, Imberti L, Sottini A, Quiros-Roldan E, Castelli F, Rossi C, Brugnoni D, Biondi A, Bettini LR, D'Angio M, Bonfanti P, Anderson MV, Saracino A, Chironna M, Di Stefano M, Fiore JR, Santantonio T, Castagnoli R, Marseglia GL, Magliocco M, Bosticardo M, Pala F, Shaw E, Matthews H, Weber SE, Xirasagar S, Barnett J, Oler AJ, Dimitrova D, Bergerson JRE, McDermott DH, Rao VK, Murphy PM, Holland SM, Lisco A, Su HC, Lionakis MS, Cohen JI, Freeman AF, Snyder TM, Lack J, Notarangelo LD. Perturbations of the T-cell receptor repertoire in response to SARS-CoV-2 in immunocompetent and immunocompromised individuals. J Allergy Clin Immunol 2024; 153:1655-1667. [PMID: 38154666 PMCID: PMC11162338 DOI: 10.1016/j.jaci.2023.12.011] [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: 05/29/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Functional T-cell responses are essential for virus clearance and long-term protection after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, whereas certain clinical factors, such as older age and immunocompromise, are associated with worse outcome. OBJECTIVE We sought to study the breadth and magnitude of T-cell responses in patients with coronavirus disease 2019 (COVID-19) and in individuals with inborn errors of immunity (IEIs) who had received COVID-19 mRNA vaccine. METHODS Using high-throughput sequencing and bioinformatics tools to characterize the T-cell receptor β repertoire signatures in 540 individuals after SARS-CoV-2 infection, 31 IEI recipients of COVID-19 mRNA vaccine, and healthy controls, we quantified HLA class I- and class II-restricted SARS-CoV-2-specific responses and also identified several HLA allele-clonotype motif associations in patients with COVID-19, including a subcohort of anti-type 1 interferon (IFN-1)-positive patients. RESULTS Our analysis revealed that elderly patients with COVID-19 with critical disease manifested lower SARS-CoV-2 T-cell clonotype diversity as well as T-cell responses with reduced magnitude, whereas the SARS-CoV-2-specific clonotypes targeted a broad range of HLA class I- and class II-restricted epitopes across the viral proteome. The presence of anti-IFN-I antibodies was associated with certain HLA alleles. Finally, COVID-19 mRNA immunization induced an increase in the breadth of SARS-CoV-2-specific clonotypes in patients with IEIs, including those who had failed to seroconvert. CONCLUSIONS Elderly individuals have impaired capacity to develop broad and sustained T-cell responses after SARS-CoV-2 infection. Genetic factors may play a role in the production of anti-IFN-1 antibodies. COVID-19 mRNA vaccines are effective in inducing T-cell responses in patients with IEIs.
Collapse
Affiliation(s)
- Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Katherine Myint-Hpu
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Boaz Palterer
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michael S Abers
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Deborah Draper
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Meng Truong
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | | | - Yu Zhang
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Lindsey B Rosen
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrew L Snow
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md; Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - Clifton L Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, Md; The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - Peter D Burbelo
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Md
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Alessandra Sottini
- Section of Microbiology, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Eugenia Quiros-Roldan
- Department of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Francesco Castelli
- Department of Infectious and Tropical Diseases, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Camillo Rossi
- Direzione Sanitaria, ASST Spedali Civili, Brescia, Italy
| | - Duilio Brugnoni
- Laboratorio Analisi Chimico-Cliniche, ASST Spedali Civili, Brescia, Italy
| | - Andrea Biondi
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Laura Rachele Bettini
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Mariella D'Angio
- Pediatric Department and Centro Tettamanti-European Reference Network on Paediatric Cancer, European Reference Network on Haematological Diseases, and European Reference Network on Hereditary Metabolic Disorders, University of Milano-Bicocca-Fondazione MBBM, Monza, Italy
| | - Paolo Bonfanti
- Department of Infectious Diseases, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Megan V Anderson
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Annalisa Saracino
- Clinic of Infectious Diseases, Azienda Ospedaliero-Universitaria Consorziale Policlinico di Bari, University of Bari, Bari, Italy
| | - Maria Chironna
- Hygiene Section, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Mariantonietta Di Stefano
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Jose Ramon Fiore
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Teresa Santantonio
- Department of Medical and Surgical Sciences, Section of Infectious Diseases, University of Foggia, Foggia, Italy
| | - Riccardo Castagnoli
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gian Luigi Marseglia
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mary Magliocco
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Elana Shaw
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Helen Matthews
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sarah E Weber
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sandhya Xirasagar
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jason Barnett
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrew J Oler
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Dimana Dimitrova
- Center for Immuno-Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md
| | - Jenna R E Bergerson
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - David H McDermott
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - V Koneti Rao
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Andrea Lisco
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michail S Lionakis
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Jeffrey I Cohen
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | - Justin Lack
- Integrated Data Sciences Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
31
|
Aguilar-Bretones M, den Hartog Y, van Dijk LLA, Malahe SRK, Dieterich M, Mora HT, Mueller YM, Koopmans MPG, Reinders MEJ, Baan CC, van Nierop GP, de Vries RD. SARS-CoV-2-specific immune responses converge in kidney disease patients and controls with hybrid immunity. NPJ Vaccines 2024; 9:93. [PMID: 38806532 PMCID: PMC11133345 DOI: 10.1038/s41541-024-00886-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/07/2024] [Indexed: 05/30/2024] Open
Abstract
Healthy individuals with hybrid immunity, due to a SARS-CoV-2 infection prior to first vaccination, have stronger immune responses compared to those who were exclusively vaccinated. However, little is known about the characteristics of antibody, B- and T-cell responses in kidney disease patients with hybrid immunity. Here, we explored differences between kidney disease patients and controls with hybrid immunity after asymptomatic or mild coronavirus disease-2019 (COVID-19). We studied the kinetics, magnitude, breadth and phenotype of SARS-CoV-2-specific immune responses against primary mRNA-1273 vaccination in patients with chronic kidney disease or on dialysis, kidney transplant recipients, and controls with hybrid immunity. Although vaccination alone is less immunogenic in kidney disease patients, mRNA-1273 induced a robust immune response in patients with prior SARS-CoV-2 infection. In contrast, kidney disease patients with hybrid immunity develop SARS-CoV-2 antibody, B- and T-cell responses that are equally strong or stronger than controls. Phenotypic analysis showed that Spike (S)-specific B-cells varied between groups in lymph node-homing and memory phenotypes, yet S-specific T-cell responses were phenotypically consistent across groups. The heterogeneity amongst immune responses in hybrid immune kidney patients warrants further studies in larger cohorts to unravel markers of long-term protection that can be used for the design of targeted vaccine regimens.
Collapse
Affiliation(s)
| | - Yvette den Hartog
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Laura L A van Dijk
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - S Reshwan K Malahe
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Marjolein Dieterich
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Héctor Tejeda Mora
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Yvonne M Mueller
- Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | | | - Rory D de Vries
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
32
|
Ramos A, Martins S, Marinho AS, Norton P, Cardoso MJ, Guimarães JT. Evaluation of SARS-CoV-2 interferon gamma release assay in BNT162b2 vaccinated healthcare workers. PLoS One 2024; 19:e0303244. [PMID: 38728294 PMCID: PMC11086832 DOI: 10.1371/journal.pone.0303244] [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: 04/21/2023] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
To predict protective immunity to SARS-CoV-2, cellular immunity seems to be more sensitive than humoral immunity. Through an Interferon-Gamma (IFN-γ) Release Assay (IGRA), we show that, despite a marked decrease in total antibodies, 94.3% of 123 healthcare workers have a positive cellular response 6 months after inoculation with the 2nd dose of BNT162b2 vaccine. Despite the qualitative relationship found, we did not observe a quantitative correlation between IFN-γ and IgG levels against SARS-CoV-2. Using stimulated whole blood from a subset of participants, we confirmed the specific T-cell response to SARS-CoV-2 by dosing elevated levels of the IL-6, IL-10 and TNF-α. Through a 20-month follow-up, we found that none of the infected participants had severe COVID-19 and that the first positive cases were only 12 months after the 2nd dose inoculation. Future studies are needed to understand if IGRA-SARS-CoV-2 can be a powerful diagnostic tool to predict future COVID-19 severe disease, guiding vaccination policies.
Collapse
Affiliation(s)
- Angélica Ramos
- Serviço de Patologia Clínica, Centro Hospitalar Universitário de São João, Porto, Portugal
- EPIUnit–Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
| | - Sandra Martins
- Serviço de Patologia Clínica, Centro Hospitalar Universitário de São João, Porto, Portugal
- EPIUnit–Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
| | - Ana Sofia Marinho
- Serviço de Patologia Clínica, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Pedro Norton
- EPIUnit–Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
- Serviço de Saúde Ocupacional, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Maria João Cardoso
- Serviço de Patologia Clínica, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - João Tiago Guimarães
- Serviço de Patologia Clínica, Centro Hospitalar Universitário de São João, Porto, Portugal
- EPIUnit–Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
- Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
33
|
González Cueto E, Edmans M, Wellens J, Cadwell K, Thompson C, Satsangi J, Wong SY. Antibody, not Cellular, Immune Responses to SARS-CoV-2 Vaccination Outperform Infection in Inflammatory Bowel Disease Patients. Inflamm Bowel Dis 2024; 30:859-862. [PMID: 37935466 DOI: 10.1093/ibd/izad265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Indexed: 11/09/2023]
Affiliation(s)
| | - Matthew Edmans
- University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, United Kingdom
| | - Judith Wellens
- University Hospitals Leuven-KU Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
- University of Oxford, Translational Gastroenterology Unit, Nuffield Department of Medicine, Oxford, United Kingdom
| | - Ken Cadwell
- Warwick Medical School- University of Warwick, Division of Biomedical Sciences, Coventry, United Kingdom
| | - Craig Thompson
- University of Pennsylvania-Perelman School of Medicine, Division of Gastroenterology, Department of Medicine, Philadelphia, United States
| | - Jack Satsangi
- University of Oxford, Translational Gastroenterology Unit, Nuffield Department of Medicine, Oxford, United Kingdom
| | - Serre-Yu Wong
- Icahn School of Medicine at Mount Sinai, The Henry D. Janowitz Division of Gastroenterology, New York, United States
| |
Collapse
|
34
|
Merli M, Costantini A, Tafuri S, Bavaro DF, Minoia C, Meli E, Luminari S, Gini G. Management of vaccinations in patients with non-Hodgkin lymphoma. Br J Haematol 2024; 204:1617-1634. [PMID: 38532527 DOI: 10.1111/bjh.19422] [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: 12/19/2023] [Revised: 03/07/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
Vaccinations are fundamental tools in preventing infectious diseases, especially in immunocompromised patients like those affected by non-Hodgkin lymphomas (NHLs). The COVID-19 pandemic made clinicians increasingly aware of the importance of vaccinations in preventing potential life-threatening SARS-CoV-2-related complications in NHL patients. However, several studies have confirmed a significant reduction in vaccine-induced immune responses after anti-CD20 monoclonal antibody treatment, thus underscoring the need for refined immunization strategies in NHL patients. In this review, we summarize the existing data about COVID-19 and other vaccine's efficacy in patients with NHL and propose multidisciplinary team-based recommendations for the management of vaccines in this specific group of patients.
Collapse
Affiliation(s)
- Michele Merli
- Division of Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Costantini
- Clinical Immunology Unit, Azienda Ospedaliero Universitaria delle Marche - Università Politecnica delle Marche, Ancona, Italy
| | - Silvio Tafuri
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Davide Fiore Bavaro
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, Aldo Moro University of Bari, Bari, Italy
| | - Carla Minoia
- Hematology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Erika Meli
- Division of Hematology, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Stefano Luminari
- Hematology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Surgical Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Guido Gini
- Clinic of Hematology, Azienda Ospedaliero Universitaria Delle Marche - Università Politecnica Delle Marche, Ancona, Italy
| |
Collapse
|
35
|
Tong S, Scott JC, Eyoh E, Werthmann DW, Stone AE, Murrell AE, Sabino-Santos G, Trinh IV, Chandra S, Elliott DH, Smira AR, Velazquez JV, Schieffelin J, Ning B, Hu T, Kolls JK, Landry SJ, Zwezdaryk KJ, Robinson JE, Gunn BM, Rabito FA, Norton EB. Altered COVID-19 immunity in children with asthma by atopic status. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100236. [PMID: 38590754 PMCID: PMC11000189 DOI: 10.1016/j.jacig.2024.100236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 04/10/2024]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes a spectrum of clinical outcomes that may be complicated by severe asthma. Antiviral immunity is often compromised in patients with asthma; however, whether this is true for SARS-CoV-2 immunity and children is unknown. Objective We aimed to evaluate SARS-CoV-2 immunity in children with asthma on the basis of infection or vaccination history and compared to respiratory syncytial viral or allergen (eg, cockroach, dust mite)-specific immunity. Methods Fifty-three children from an urban asthma study were evaluated for medical history, lung function, and virus- or allergen-specific immunity using antibody or T-cell assays. Results Polyclonal antibody responses to spike were observed in most children from infection and/or vaccination history. Children with atopic asthma or high allergen-specific IgE, particularly to dust mites, exhibited reduced seroconversion, antibody magnitude, and SARS-CoV-2 virus neutralization after SARS-CoV-2 infection or vaccination. TH1 responses to SARS-CoV-2 and respiratory syncytial virus correlated with antigen-respective IgG. Cockroach-specific T-cell activation as well as IL-17A and IL-21 cytokines negatively correlated with SARS-CoV-2 antibodies and effector functions, distinct from total and dust mite IgE. Allergen-specific IgE and lack of vaccination were associated with recent health care utilization. Reduced lung function (forced expiratory volume in 1 second ≤ 80%) was independently associated with (SARS-CoV-2) peptide-induced cytokines, including IL-31, whereas poor asthma control was associated with cockroach-specific cytokine responses. Conclusion Mechanisms underpinning atopic and nonatopic asthma may complicate the development of memory to SARS-CoV-2 infection or vaccination and lead to a higher risk of repeated infection in these children.
Collapse
Affiliation(s)
- Sherry Tong
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Jordan C. Scott
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Enwono Eyoh
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Derek W. Werthmann
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, La
| | - Addison E. Stone
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Amelie E. Murrell
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Gilberto Sabino-Santos
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Ivy V. Trinh
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Sruti Chandra
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Debra H. Elliott
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Ashley R. Smira
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Jalene V. Velazquez
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| | - John Schieffelin
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Bo Ning
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, La
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, La
| | - Tony Hu
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, La
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, La
| | - Jay K. Kolls
- Department of Medicine, Tulane University School of Medicine, New Orleans, La
| | - Samuel J. Landry
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, La
| | - Kevin J. Zwezdaryk
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - James E. Robinson
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Bronwyn M. Gunn
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| | - Felicia A. Rabito
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, La
| | - Elizabeth B. Norton
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| |
Collapse
|
36
|
Reemann L, Kneidinger N, Sczepanski B, Koczulla AR. COVID-19 in Lung Transplant Recipients: A Report on 10 Recent Cases. Viruses 2024; 16:709. [PMID: 38793590 PMCID: PMC11126037 DOI: 10.3390/v16050709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Due to immunosuppression, transplant recipients are at higher risk of infections with SARS-CoV-2 and worse clinical outcomes than immunocompetent hosts. Furthermore, lung transplant patients represent a special group among solid organ recipients, since pneumonia is the main manifestation of COVID-19. However, data on the course of disease and the changes in morbidity and mortality during the course of the pandemic are limited. In our pulmonary rehabilitation clinic, we treat patients shortly after lung transplant as well as long-term transplant patients. Over the last almost 4 years of pandemic, we witnessed several COVID-19 infections in lung transplant patients in our clinic as well as patients who acquired an infection beforehand. In this paper, we aim at retrospectively describing a series of recent COVID-19 cases in our clinic, looking at the clinical course of disease and outcomes in lung transplant patients.
Collapse
Affiliation(s)
- Lea Reemann
- Institute for Pulmonary Rehabilitation Research, Schoen Klinik Berchtesgadener Land, 83471 Schoenau am Koenigssee, Germany; (B.S.); (A.R.K.)
| | - Nikolaus Kneidinger
- Department of Medicine V, Comprehensive Pneumology Center Munich (CPC-M), German Center for Lung Research (DZL), Ludwig-Maximilians University (LMU) University Hospital, 81377 Munich, Germany;
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Bernd Sczepanski
- Institute for Pulmonary Rehabilitation Research, Schoen Klinik Berchtesgadener Land, 83471 Schoenau am Koenigssee, Germany; (B.S.); (A.R.K.)
| | - Andreas Rembert Koczulla
- Institute for Pulmonary Rehabilitation Research, Schoen Klinik Berchtesgadener Land, 83471 Schoenau am Koenigssee, Germany; (B.S.); (A.R.K.)
- Department of Pulmonary Rehabilitation, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-University of Marburg, 35043 Marburg, Germany
- Teaching Hospital, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| |
Collapse
|
37
|
Zhang N, Wang Z, Li H, Chen K, Wang HS, Shao JB, Jiang SY, Zhai XW, Jiang H. Immunophenotype of lymphocytes and real-world outcome of COVID-19 infection in children with hematology and oncology. BMC Cancer 2024; 24:538. [PMID: 38678181 PMCID: PMC11056048 DOI: 10.1186/s12885-024-12262-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Patients with immunocompromise were suspected to encounter a high risk for severe coronavirus disease 2019 (COVID-19) infection on early period; however, data is lacking nowadays and immune response remain unclear. METHODS In this retrospective study, internet questionnaire survey and medical records were acquired in pediatric hematology oncology patients. Clinical severity, immunological characteristics, and outcomes were analyzed from December 1, 2022 to January 31, 2023 at the 3rd year of pandemic in China. RESULTS A total of 306 patients were included, with 21 patients (6.9%) asymptomatic, 262 (85.6%) mild severity, 17 (5.6%) moderate severity, 5 (1.6%) severe severity, and 1 (0.3%) critical severity. Seventy-eight (25.5%) patients were on intensive chemotherapy, and 32.0% children were on maintenance chemotherapy. Delays in cancer therapy occurred in 86.7% patients. Univariable analysis revealed active chemotherapy (P < 0.0001), long duration of symptom (P < 0.0001), low lymphocytes count (P = 0.095), low CD3 + and CD8 + T cell count (P = 0.013, P = 0.022), high percentage of CD4 + TCM (P = 0.016), and low percentage of transitional B cells (P = 0.045) were high risk factors for severe COVID-19 infection. Cox regression model showed that the absolute lymphocytes count (P = 0.027) and long duration of symptom (P = 0.002) were the independent factors for severity. Patients with CD8 + dominant and B cell depletion subtype wasn't related with severity, but had higher percentage of CD8 + effector memory T cells (TEM) and terminally differentiated effector memory T cells (TEMRA) (P < 0.001, P < 0.001), and a longer COVID-19 duration (P = 0.045). CONCLUSION The severity was relatively mild in children with immunodeficiencies in the third year of COVID-19 pandemic. Low lymphocyte count and long duration of symptom were the independent risk factors with COVID-19 severity. Delays in cancer care remain a major concern and the long outcome is pending.
Collapse
Affiliation(s)
- Na Zhang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Zhen Wang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Hong Li
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Kai Chen
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Hong-Sheng Wang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Jing-Bo Shao
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Sha-Yi Jiang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Xiao-Wen Zhai
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Hui Jiang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China.
| |
Collapse
|
38
|
Chi H, Pepper M, Thomas PG. Principles and therapeutic applications of adaptive immunity. Cell 2024; 187:2052-2078. [PMID: 38670065 PMCID: PMC11177542 DOI: 10.1016/j.cell.2024.03.037] [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: 01/02/2024] [Revised: 03/01/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Adaptive immunity provides protection against infectious and malignant diseases. These effects are mediated by lymphocytes that sense and respond with targeted precision to perturbations induced by pathogens and tissue damage. Here, we review key principles underlying adaptive immunity orchestrated by distinct T cell and B cell populations and their extensions to disease therapies. We discuss the intracellular and intercellular processes shaping antigen specificity and recognition in immune activation and lymphocyte functions in mediating effector and memory responses. We also describe how lymphocytes balance protective immunity against autoimmunity and immunopathology, including during immune tolerance, response to chronic antigen stimulation, and adaptation to non-lymphoid tissues in coordinating tissue immunity and homeostasis. Finally, we discuss extracellular signals and cell-intrinsic programs underpinning adaptive immunity and conclude by summarizing key advances in vaccination and engineering adaptive immune responses for therapeutic interventions. A deeper understanding of these principles holds promise for uncovering new means to improve human health.
Collapse
Affiliation(s)
- Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Marion Pepper
- Department of Immunology, University of Washington, Seattle, WA, USA.
| | - Paul G Thomas
- Department of Host-Microbe Interactions and Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
39
|
Wang J, Lu W, Zhang J, Du Y, Fang M, Zhang A, Sungcad G, Chon S, Xing J. Loss of TRIM29 mitigates viral myocarditis by attenuating PERK-driven ER stress response in male mice. Nat Commun 2024; 15:3481. [PMID: 38664417 PMCID: PMC11045800 DOI: 10.1038/s41467-024-44745-x] [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: 05/23/2023] [Accepted: 12/29/2023] [Indexed: 04/28/2024] Open
Abstract
Viral myocarditis, an inflammatory disease of the myocardium, is a significant cause of sudden death in children and young adults. The current coronavirus disease 19 pandemic emphasizes the need to understand the pathogenesis mechanisms and potential treatment strategies for viral myocarditis. Here, we found that TRIM29 was highly induced by cardiotropic viruses and promoted protein kinase RNA-like endoplasmic reticulum kinase (PERK)-mediated endoplasmic reticulum (ER) stress, apoptosis, and reactive oxygen species (ROS) responses that promote viral replication in cardiomyocytes in vitro. TRIM29 deficiency protected mice from viral myocarditis by promoting cardiac antiviral functions and reducing PERK-mediated inflammation and immunosuppressive monocytic myeloid-derived suppressor cells (mMDSC) in vivo. Mechanistically, TRIM29 interacted with PERK to promote SUMOylation of PERK to maintain its stability, thereby promoting PERK-mediated signaling pathways. Finally, we demonstrated that the PERK inhibitor GSK2656157 mitigated viral myocarditis by disrupting the TRIM29-PERK connection, thereby bolstering cardiac function, enhancing cardiac antiviral responses, and curbing inflammation and immunosuppressive mMDSC in vivo. Our findings offer insight into how cardiotropic viruses exploit TRIM29-regulated PERK signaling pathways to instigate viral myocarditis, suggesting that targeting the TRIM29-PERK axis could mitigate disease severity.
Collapse
Affiliation(s)
- Junying Wang
- Department of Surgery and Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston Methodist, Houston, TX, 77030, USA
| | - Wenting Lu
- Department of Surgery and Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston Methodist, Houston, TX, 77030, USA
| | - Jerry Zhang
- Department of Surgery and Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston Methodist, Houston, TX, 77030, USA
| | - Yong Du
- Department of Surgery and Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston Methodist, Houston, TX, 77030, USA
| | - Mingli Fang
- Department of Surgery and Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston Methodist, Houston, TX, 77030, USA
| | - Ao Zhang
- Department of Surgery and Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston Methodist, Houston, TX, 77030, USA
| | - Gabriel Sungcad
- Department of Surgery and Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston Methodist, Houston, TX, 77030, USA
| | - Samantha Chon
- Department of Surgery and Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston Methodist, Houston, TX, 77030, USA
| | - Junji Xing
- Department of Surgery and Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston Methodist, Houston, TX, 77030, USA.
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Methodist, Houston, TX, 77030, USA.
- Department of Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA.
| |
Collapse
|
40
|
Kempaiah P, Libertin CR, Chitale RA, Naeyma I, Pleqi V, Sheele JM, Iandiorio MJ, Hoogesteijn AL, Caulfield TR, Rivas AL. Decoding Immuno-Competence: A Novel Analysis of Complete Blood Cell Count Data in COVID-19 Outcomes. Biomedicines 2024; 12:871. [PMID: 38672225 PMCID: PMC11048687 DOI: 10.3390/biomedicines12040871] [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: 02/10/2024] [Revised: 03/14/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND While 'immuno-competence' is a well-known term, it lacks an operational definition. To address this omission, this study explored whether the temporal and structured data of the complete blood cell count (CBC) can rapidly estimate immuno-competence. To this end, one or more ratios that included data on all monocytes, lymphocytes and neutrophils were investigated. MATERIALS AND METHODS Longitudinal CBC data collected from 101 COVID-19 patients (291 observations) were analyzed. Dynamics were estimated with several approaches, which included non-structured (the classic CBC format) and structured data. Structured data were assessed as complex ratios that capture multicellular interactions among leukocytes. In comparing survivors with non-survivors, the hypothesis that immuno-competence may exhibit feedback-like (oscillatory or cyclic) responses was tested. RESULTS While non-structured data did not distinguish survivors from non-survivors, structured data revealed immunological and statistical differences between outcomes: while survivors exhibited oscillatory data patterns, non-survivors did not. In survivors, many variables (including IL-6, hemoglobin and several complex indicators) showed values above or below the levels observed on day 1 of the hospitalization period, displaying L-shaped data distributions (positive kurtosis). In contrast, non-survivors did not exhibit kurtosis. Three immunologically defined data subsets included only survivors. Because information was based on visual patterns generated in real time, this method can, potentially, provide information rapidly. DISCUSSION The hypothesis that immuno-competence expresses feedback-like loops when immunological data are structured was not rejected. This function seemed to be impaired in immuno-suppressed individuals. While this method rapidly informs, it is only a guide that, to be confirmed, requires additional tests. Despite this limitation, the fact that three protective (survival-associated) immunological data subsets were observed since day 1 supports many clinical decisions, including the early and personalized prognosis and identification of targets that immunomodulatory therapies could pursue. Because it extracts more information from the same data, structured data may replace the century-old format of the CBC.
Collapse
Affiliation(s)
- Prakasha Kempaiah
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (P.K.); (V.P.)
| | | | - Rohit A. Chitale
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Islam Naeyma
- Department of Neuroscience, Division of QHS Computational Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (I.N.); (T.R.C.)
| | - Vasili Pleqi
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (P.K.); (V.P.)
| | | | - Michelle J. Iandiorio
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA;
| | | | - Thomas R. Caulfield
- Department of Neuroscience, Division of QHS Computational Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (I.N.); (T.R.C.)
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ariel L. Rivas
- Center for Global Health, Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
41
|
Fumagalli V, Ravà M, Marotta D, Di Lucia P, Bono EB, Giustini L, De Leo F, Casalgrandi M, Monteleone E, Mouro V, Malpighi C, Perucchini C, Grillo M, De Palma S, Donnici L, Marchese S, Conti M, Muramatsu H, Perlman S, Pardi N, Kuka M, De Francesco R, Bianchi ME, Guidotti LG, Iannacone M. Antibody-independent protection against heterologous SARS-CoV-2 challenge conferred by prior infection or vaccination. Nat Immunol 2024; 25:633-643. [PMID: 38486021 PMCID: PMC11003867 DOI: 10.1038/s41590-024-01787-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/13/2024] [Indexed: 04/11/2024]
Abstract
Vaccines have reduced severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) morbidity and mortality, yet emerging variants challenge their effectiveness. The prevailing approach to updating vaccines targets the antibody response, operating under the presumption that it is the primary defense mechanism following vaccination or infection. This perspective, however, can overlook the role of T cells, particularly when antibody levels are low or absent. Here we show, through studies in mouse models lacking antibodies but maintaining functional B cells and lymphoid organs, that immunity conferred by prior infection or mRNA vaccination can protect against SARS-CoV-2 challenge independently of antibodies. Our findings, using three distinct models inclusive of a novel human/mouse ACE2 hybrid, highlight that CD8+ T cells are essential for combating severe infections, whereas CD4+ T cells contribute to managing milder cases, with interferon-γ having an important function in this antibody-independent defense. These findings highlight the importance of T cell responses in vaccine development, urging a broader perspective on protective immunity beyond just antibodies.
Collapse
Affiliation(s)
- Valeria Fumagalli
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Micol Ravà
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Davide Marotta
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Pietro Di Lucia
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Elisa B Bono
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Leonardo Giustini
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica De Leo
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | | | - Violette Mouro
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Malpighi
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Perucchini
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Grillo
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Sara De Palma
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Charles River Laboratories, Calco, Italy
| | - Lorena Donnici
- Istituto Nazionale di Genetica Molecolare (INGM) 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Silvia Marchese
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Matteo Conti
- Istituto Nazionale di Genetica Molecolare (INGM) 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mirela Kuka
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Raffaele De Francesco
- Istituto Nazionale di Genetica Molecolare (INGM) 'Romeo ed Enrica Invernizzi', Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Marco E Bianchi
- Vita-Salute San Raffaele University, Milan, Italy.
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Luca G Guidotti
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Matteo Iannacone
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
42
|
Chen F, Zhang M, Yang F, Wang L, Liu J, Liu J, Pang Y. Dual-Antigen-Displaying Nanovaccines Elicit Synergistic Immunoactivation for Treating Cancer and Preventing Infectious Complications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307748. [PMID: 38037689 DOI: 10.1002/smll.202307748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/30/2023] [Indexed: 12/02/2023]
Abstract
As one of the most common complications, infection causes the majority of mortality in cancer patients. However, therapeutic strategies that can simultaneously suppress tumors and protect patients from infection have been rarely reported. Here, the use of dual-antigen-displaying nanovaccines (DADNs) is described to elicit synergistic immunoactivation for treating cancer and preventing infectious complications. DADNs are prepared by wrapping immunoadjuvant-loaded nanoparticles with a hybrid coating, which is fused from cell membranes that are separately genetically engineered to express tumor and infectious pathogenic antigens. Due to the presence of a dual-antigen combination, DADNs are able to promote the maturation of dendritic cells and more importantly to trigger cross-presentation of both combined antigens. During in vivo investigations, we find that DADNs can reverse immunosuppression by stimulating tumor-associated antigen-specific T-cell responses, resulting in significantly delayed tumor growth in mice. These nanovaccines also elicit effective protective immunity against tumor challenges and induce robust production of pathogenic antigen-specific immunoglobulin G antibody in a prophylactic study. This work offers a unique approach to develop dual-mode vaccines, which are promising for synchronously treating cancer and preventing infection.
Collapse
Affiliation(s)
- Fangjie Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, Shandong, 250117, China
| | - Mengmeng Zhang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Fengmin Yang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lu Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Junqiu Liu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yan Pang
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| |
Collapse
|
43
|
Huang L, Tong X, Cui J, Du X, Liao Y, Tan X, Ju Y, Zhong X, Zhou W, Xu X, Li Y. Recurrent and persistent fever after SARS-CoV-2 infection in patients with follicular lymphoma: A case series. Int J Infect Dis 2024; 141:106973. [PMID: 38395220 DOI: 10.1016/j.ijid.2024.02.016] [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: 12/13/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Although persistent or recurrent COVID-19 infection is well described in some immunosuppressed patient cohort, to date, there have been no reports of this phenomenon in the context of repeatedly negative SARS-CoV-2 testing in the upper respiratory tract. We reported six patients with follicular lymphoma who developed recurrent symptomatic COVID-19 infection. They tested persistently negative for SARS-CoV-2 on pharyngeal swabs and ultimately confirmed by bronchoalveolar lavage fluid metagenomics next-generation sequencing. All six patients presented with lymphopenia and B-cell depletion, and five of them received the anti-cluster of differentiation 20 treatment in the last year. Persistent fever was the most common symptom and bilateral ground-glass opacities were the primary pattern on chest computed tomography. A relatively long course of unnecessary and ineffective antibacterial and/or antifungal treatments was administered until the definitive diagnosis. Persistent fever subsided rapidly with nirmatrelvir/ritonavir treatment. Our case highlighted that recurrent COVID-19 infection should be suspected in immunocompromised patients with persistent fever despite negative pharyngeal swabs, and urgent bronchoalveolar lavage fluid testing is necessary. Treatment with nirmatrelvir/ritonavir appeared to be very effective in these patients.
Collapse
Affiliation(s)
- Lixue Huang
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Xunliang Tong
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Jia Cui
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Xiaoman Du
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Yixuan Liao
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Xiaoming Tan
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Yang Ju
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Xuefeng Zhong
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Wei Zhou
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Xiaomao Xu
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Yanming Li
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China.
| |
Collapse
|
44
|
Camelo ALM, Zamora Obando HR, Rocha I, Dias AC, Mesquita ADS, Simionato AVC. COVID-19 and Comorbidities: What Has Been Unveiled by Metabolomics? Metabolites 2024; 14:195. [PMID: 38668323 PMCID: PMC11051775 DOI: 10.3390/metabo14040195] [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: 01/18/2024] [Revised: 03/14/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
The COVID-19 pandemic has brought about diverse impacts on the global population. Individuals with comorbidities were more susceptible to the severe symptoms caused by the virus. Within the crisis scenario, metabolomics represents a potential area of science capable of providing relevant information for understanding the metabolic pathways associated with the intricate interaction between the viral disease and previous comorbidities. This work aims to provide a comprehensive description of the scientific production pertaining to metabolomics within the specific context of COVID-19 and comorbidities, while highlighting promising areas for exploration by those interested in the subject. In this review, we highlighted the studies of metabolomics that indicated a variety of metabolites associated with comorbidities and COVID-19. Furthermore, we observed that the understanding of the metabolic processes involved between comorbidities and COVID-19 is limited due to the urgent need to report disease outcomes in individuals with comorbidities. The overlap of two or more comorbidities associated with the severity of COVID-19 hinders the comprehension of the significance of each condition. Most identified studies are observational, with a restricted number of patients, due to challenges in sample collection amidst the emergent situation.
Collapse
Affiliation(s)
- André Luiz Melo Camelo
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Hans Rolando Zamora Obando
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Isabela Rocha
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Aline Cristina Dias
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Alessandra de Sousa Mesquita
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
| | - Ana Valéria Colnaghi Simionato
- Laboratory of Analysis of Biomolecules Tiselius, Department of Analytical Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil; (A.L.M.C.); (H.R.Z.O.); (I.R.); (A.C.D.); (A.d.S.M.)
- National Institute of Science and Technology for Bioanalytics—INCTBio, Institute of Chemistry, Universidade Estadual de (UNICAMP), Campinas 13083-970, São Paulo, Brazil
| |
Collapse
|
45
|
Coulon PG, Prakash S, Dhanushkodi NR, Srivastava R, Zayou L, Tifrea DF, Edwards RA, Figueroa CJ, Schubl SD, Hsieh L, Nesburn AB, Kuppermann BD, Bahraoui E, Vahed H, Gil D, Jones TM, Ulmer JB, BenMohamed L. High frequencies of alpha common cold coronavirus/SARS-CoV-2 cross-reactive functional CD4 + and CD8 + memory T cells are associated with protection from symptomatic and fatal SARS-CoV-2 infections in unvaccinated COVID-19 patients. Front Immunol 2024; 15:1343716. [PMID: 38605956 PMCID: PMC11007208 DOI: 10.3389/fimmu.2024.1343716] [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: 11/24/2023] [Accepted: 03/08/2024] [Indexed: 04/13/2024] Open
Abstract
Background Cross-reactive SARS-CoV-2-specific memory CD4+ and CD8+ T cells are present in up to 50% of unexposed, pre-pandemic, healthy individuals (UPPHIs). However, the characteristics of cross-reactive memory CD4+ and CD8+ T cells associated with subsequent protection of asymptomatic coronavirus disease 2019 (COVID-19) patients (i.e., unvaccinated individuals who never develop any COVID-19 symptoms despite being infected with SARS-CoV-2) remains to be fully elucidated. Methods This study compares the antigen specificity, frequency, phenotype, and function of cross-reactive memory CD4+ and CD8+ T cells between common cold coronaviruses (CCCs) and SARS-CoV-2. T-cell responses against genome-wide conserved epitopes were studied early in the disease course in a cohort of 147 unvaccinated COVID-19 patients who were divided into six groups based on the severity of their symptoms. Results Compared to severely ill COVID-19 patients and patients with fatal COVID-19 outcomes, the asymptomatic COVID-19 patients displayed significantly: (i) higher rates of co-infection with the 229E alpha species of CCCs (α-CCC-229E); (ii) higher frequencies of cross-reactive functional CD134+CD137+CD4+ and CD134+CD137+CD8+ T cells that cross-recognized conserved epitopes from α-CCCs and SARS-CoV-2 structural, non-structural, and accessory proteins; and (iii) lower frequencies of CCCs/SARS-CoV-2 cross-reactive exhausted PD-1+TIM3+TIGIT+CTLA4+CD4+ and PD-1+TIM3+TIGIT+CTLA4+CD8+ T cells, detected both ex vivo and in vitro. Conclusions These findings (i) support a crucial role of functional, poly-antigenic α-CCCs/SARS-CoV-2 cross-reactive memory CD4+ and CD8+ T cells, induced following previous CCCs seasonal exposures, in protection against subsequent severe COVID-19 disease and (ii) provide critical insights into developing broadly protective, multi-antigen, CD4+, and CD8+ T-cell-based, universal pan-Coronavirus vaccines capable of conferring cross-species protection.
Collapse
Affiliation(s)
- Pierre-Gregoire Coulon
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, United States
| | - Swayam Prakash
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, United States
| | - Nisha R. Dhanushkodi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, United States
| | - Ruchi Srivastava
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, United States
| | - Latifa Zayou
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, United States
| | - Delia F. Tifrea
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, United States
| | - Robert A. Edwards
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, United States
| | - Cesar J. Figueroa
- Department of Surgery, Divisions of Trauma, Burns and Critical Care, School of Medicine, University of California Irvine, Irvine, CA, United States
| | - Sebastian D. Schubl
- Department of Surgery, Divisions of Trauma, Burns and Critical Care, School of Medicine, University of California Irvine, Irvine, CA, United States
| | - Lanny Hsieh
- Department of Medicine, Division of Infectious Diseases and Hospitalist Program, School of Medicine, University of California Irvine, Irvine, CA, United States
| | - Anthony B. Nesburn
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, United States
| | - Baruch D. Kuppermann
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, United States
| | | | - Hawa Vahed
- Department of Vaccines and Immunotherapies, TechImmune, LLC, University Lab Partners, Irvine, CA, United States
| | - Daniel Gil
- Department of Vaccines and Immunotherapies, TechImmune, LLC, University Lab Partners, Irvine, CA, United States
| | - Trevor M. Jones
- Department of Vaccines and Immunotherapies, TechImmune, LLC, University Lab Partners, Irvine, CA, United States
| | - Jeffrey B. Ulmer
- Department of Vaccines and Immunotherapies, TechImmune, LLC, University Lab Partners, Irvine, CA, United States
| | - Lbachir BenMohamed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, University of California Irvine, School of Medicine, Irvine, CA, United States
- Université Paul Sabatier, Infinity, Inserm, Toulouse, France
- Department of Vaccines and Immunotherapies, TechImmune, LLC, University Lab Partners, Irvine, CA, United States
- Institute for Immunology, The University of California Irvine, School of Medicine, Irvine, CA, United States
| |
Collapse
|
46
|
Terzoli S, Marzano P, Cazzetta V, Piazza R, Sandrock I, Ravens S, Tan L, Prinz I, Balin S, Calvi M, Carletti A, Cancellara A, Coianiz N, Franzese S, Frigo A, Voza A, Calcaterra F, Di Vito C, Della Bella S, Mikulak J, Mavilio D. Expansion of memory Vδ2 T cells following SARS-CoV-2 vaccination revealed by temporal single-cell transcriptomics. NPJ Vaccines 2024; 9:63. [PMID: 38509155 PMCID: PMC10954735 DOI: 10.1038/s41541-024-00853-9] [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: 01/20/2023] [Accepted: 03/05/2024] [Indexed: 03/22/2024] Open
Abstract
γδ T cells provide rapid cellular immunity against pathogens. Here, we conducted matched single-cell RNA-sequencing and γδ-TCR-sequencing to delineate the molecular changes in γδ T cells during a longitudinal study following mRNA SARS-CoV-2 vaccination. While the first dose of vaccine primes Vδ2 T cells, it is the second administration that significantly boosts their immune response. Specifically, the second vaccination uncovers memory features of Vδ2 T cells, shaped by the induction of AP-1 family transcription factors and characterized by a convergent central memory signature, clonal expansion, and an enhanced effector potential. This temporally distinct effector response of Vδ2 T cells was also confirmed in vitro upon stimulation with SARS-CoV-2 spike-peptides. Indeed, the second challenge triggers a significantly higher production of IFNγ by Vδ2 T cells. Collectively, our findings suggest that mRNA SARS-CoV-2 vaccination might benefit from the establishment of long-lasting central memory Vδ2 T cells to confer protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Sara Terzoli
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
| | - Paolo Marzano
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Valentina Cazzetta
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School (MHH), Hannover, Germany
| | - Sarina Ravens
- Institute of Immunology, Hannover Medical School (MHH), Hannover, Germany
| | - Likai Tan
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School (MHH), Hannover, Germany
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Balin
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Michela Calvi
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Anna Carletti
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Assunta Cancellara
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Nicolò Coianiz
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Sara Franzese
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Alessandro Frigo
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Antonio Voza
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
- Department of Biomedical Unit, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Francesca Calcaterra
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Clara Di Vito
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Silvia Della Bella
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Joanna Mikulak
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy.
| | - Domenico Mavilio
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy.
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| |
Collapse
|
47
|
Calderón-Parra J, Gutiérrez-Villanueva A, Ronda-Roca G, Jimenez MLM, de la Torre H, Ródenas-Baquero M, Paniura-Pinedo M, Lozano-Llano C, Pintos-Pascual I, Fernández-Cruz A, Ramos-Martínez A, Muñez-Rubio E. Efficacy and safety of antiviral plus anti-spike monoclonal antibody combination therapy vs. monotherapy for high-risk immunocompromised patients with mild-to-moderate SARS-CoV2 infection during the Omicron era: A prospective cohort study. Int J Antimicrob Agents 2024; 63:107095. [PMID: 38244814 DOI: 10.1016/j.ijantimicag.2024.107095] [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: 09/05/2023] [Revised: 12/21/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
INTRODUCTION Antivirals and monoclonal antibodies lower the risk of progression in immunocompromised patients. However, combination therapy with both types of agents has not been studied. PATIENTS AND METHODS This was a single-centre, prospective, cohort study. All immunocompromised patients who received treatment for mild-to-moderate COVID-19 from 1 January 2022 to 30 October 2022 were enrolled. The primary endpoint was COVID-19 progression at 90 days, defined as hospital admission or death due to COVID-19 and/or seronegative persistent COVID-19. RESULTS A total of 304 patients were included: 43 patients (14.1%) received sotrovimab plus a direct-acting antiviral, and 261 (85.9%) received monotherapy. Primary outcome occurred more frequently after monotherapy (4.6% vs. 0%, P=0.154). Among patients with anti-spike immunoglobulin G (anti-S IgG) titre <750 BAU/mL, COVID-19 progression was more common after monotherapy (23.9% vs. 0%, P=0.001), including more frequent COVID-related admission (15.2% vs. 0%, P=0.014) and seronegative persistent COVID-19 (10.9% vs. 0%, P=0.044). Combination therapy was associated with lower risk of progression (odds ratio [OR] 0.08, 95% confidence interval [95% CI] 0.01-0.64). Anti-S IgG titre <750 BAU/mL and previous anti-CD20 were associated with higher risk of progression (OR 13.70, 95% CI 2.77-67.68; and OR 3.05, 95% CI 1.20-10.94, respectively). CONCLUSIONS In immunocompromised patients, combination therapy with sotrovimab plus an antiviral may be more effective than monotherapy for SARS-CoV2.
Collapse
Affiliation(s)
- Jorge Calderón-Parra
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain; Research Institute Puerta de Hierro-Segovia de Arana (IDIPHSA), Majadahonda, Spain
| | - Andrea Gutiérrez-Villanueva
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain; Research Institute Puerta de Hierro-Segovia de Arana (IDIPHSA), Majadahonda, Spain.
| | - Gerard Ronda-Roca
- Pharmacology department, University Hospital Puerta de Hierro, Majadahonda, Spain
| | | | - Helena de la Torre
- Emergency department, University Hospital Puerta de Hierro, Majadahonda, Spain
| | - María Ródenas-Baquero
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain
| | - María Paniura-Pinedo
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain
| | - Carla Lozano-Llano
- Pharmacy department, University Hospital Puerta de Hierro, Majadahonda, Spain
| | - Ilduara Pintos-Pascual
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain
| | - Ana Fernández-Cruz
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain; Research Institute Puerta de Hierro-Segovia de Arana (IDIPHSA), Majadahonda, Spain; Autónoma University of Madrid, Spain
| | - Antonio Ramos-Martínez
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain; Research Institute Puerta de Hierro-Segovia de Arana (IDIPHSA), Majadahonda, Spain; Autónoma University of Madrid, Spain
| | - Elena Muñez-Rubio
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain; Research Institute Puerta de Hierro-Segovia de Arana (IDIPHSA), Majadahonda, Spain
| |
Collapse
|
48
|
Ying B, Darling TL, Desai P, Liang CY, Dmitriev IP, Soudani N, Bricker T, Kashentseva EA, Harastani H, Raju S, Liu M, Schmidt AG, Curiel DT, Boon ACM, Diamond MS. Mucosal vaccine-induced cross-reactive CD8 + T cells protect against SARS-CoV-2 XBB.1.5 respiratory tract infection. Nat Immunol 2024; 25:537-551. [PMID: 38337035 PMCID: PMC10907304 DOI: 10.1038/s41590-024-01743-x] [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: 07/13/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
A nasally delivered chimpanzee adenoviral-vectored severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine (ChAd-SARS-CoV-2-S) is currently used in India (iNCOVACC). Here, we update this vaccine by creating ChAd-SARS-CoV-2-BA.5-S, which encodes a prefusion-stabilized BA.5 spike protein. Whereas serum neutralizing antibody responses induced by monovalent or bivalent adenoviral vaccines were poor against the antigenically distant XBB.1.5 strain and insufficient to protect in passive transfer experiments, mucosal antibody and cross-reactive memory T cell responses were robust, and protection was evident against WA1/2020 D614G and Omicron variants BQ.1.1 and XBB.1.5 in mice and hamsters. However, depletion of memory CD8+ T cells before XBB.1.5 challenge resulted in loss of protection against upper and lower respiratory tract infection. Thus, nasally delivered vaccines stimulate mucosal immunity against emerging SARS-CoV-2 strains, and cross-reactive memory CD8+ T cells mediate protection against lung infection by antigenically distant strains in the setting of low serum levels of cross-reactive neutralizing antibodies.
Collapse
Affiliation(s)
- Baoling Ying
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Tamarand L Darling
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Pritesh Desai
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Chieh-Yu Liang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Igor P Dmitriev
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nadia Soudani
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Traci Bricker
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Elena A Kashentseva
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Houda Harastani
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Saravanan Raju
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Meizi Liu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Aaron G Schmidt
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - David T Curiel
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Adrianus C M Boon
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
49
|
Gong IY, Prica A, Ante Z, Calzavara A, Krzyzanowska MK, Singh S, Suleman A, Cheung MC, Crump M. Indolent lymphoma care delivery and outcomes during the COVID-19 pandemic in Ontario, Canada. Br J Haematol 2024; 204:805-814. [PMID: 37886835 DOI: 10.1111/bjh.19166] [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: 07/29/2023] [Revised: 09/26/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023]
Abstract
The treatment pattern and outcomes in patients with indolent B-cell lymphoma treated during the coronavirus disease 2019 (COVID-19) pandemic period compared to the prepandemic period are unclear. This was a retrospective population-based study using administrative databases in Ontario, Canada (follow-up to 31 March 2022). The primary outcome was treatment pattern; secondary outcomes were death, toxicities, healthcare utilization (emergency department [ED] visit, hospitalization) and SARS-CoV-2 outcomes. Adjusted hazard ratios (aHR) from Cox proportional hazards models were used to estimate associations. We identified 4143 patients (1079 pandemic, 3064 prepandemic), with a median age of 69 years. In both time periods, bendamustine (B) + rituximab (BR) was the most frequently prescribed regimen. During the pandemic, fewer patients received R maintenance or completed the full 2-year course (aHR 0.81, 95% CI 0.71-0.92, p = 0.001). Patients treated during the pandemic had less healthcare utilization (ED visit aHR 0.77, 95% CI 0.68, 0.88, p < 0.0001; hospitalization aHR 0.81, 95% CI 0.70-0.94, p = 0.0067) and complications (infection aHR 0.69, 95% CI 0.57-0.82, p < 0.0001; febrile neutropenia aHR 0.66, 95% CI 0.47-0.94, p = 0.020), with no difference in death. Independent of vaccination, active rituximab use was associated with a higher risk of COVID-19 complications. Despite similar front-line regimen use, healthcare utilization and admissions for infection were less in the pandemic cohort.
Collapse
Affiliation(s)
- Inna Y Gong
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anca Prica
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Zharmaine Ante
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Andrew Calzavara
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
| | - Monika K Krzyzanowska
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Simron Singh
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Sunnybrook Odette Cancer Centre, Toronto, Ontario, Canada
| | - Adam Suleman
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Matthew C Cheung
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Sunnybrook Odette Cancer Centre, Toronto, Ontario, Canada
| | - Michael Crump
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Giles ML, Flanagan KL. COVID-19 vaccination: are more jabs needed or are we now immune? Intern Med J 2024; 54:368-373. [PMID: 38414215 DOI: 10.1111/imj.16341] [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: 08/14/2023] [Accepted: 01/12/2024] [Indexed: 02/29/2024]
Abstract
As the COVID-19 pandemic has progressed, it has become apparent that COVID-19 vaccination has limited impact on SAR-CoV-2 transmission and provides only short-term protection against acquiring infection, but more robust protection against severe disease and death. As a result, vaccinated people remain susceptible to SARS-CoV-2 infection but are less likely to experience severe outcomes. Studies show that immunity derived from the combination of vaccination and natural infection, so-called hybrid immunity, is superior to that provided by vaccination or natural infection alone. Since most Australian adults have received three or more doses of COVID-19 vaccines and >70% have also been infected with SARS-CoV-2, we now have a population with high levels of hybrid immunity. This was mostly achieved by receiving original Wuhan strain vaccines and then experiencing Omicron strain infections. The original Wuhan strain of SARS-CoV-2 has now disappeared and been replaced with Omicron-lineage variants globally. The predominance of the Omicron strain initially led to the development of bivalent vaccines containing both the Wuhan strain and Omicron variants. Currently, vaccines containing the original Wuhan strain of spike protein are being phased out, and new COVID-19 vaccines based exclusively on the Omicron strain XBB have become available in Australia. This article explores the question of whether further doses will be required from 2024 onwards and, if so, who should receive them?
Collapse
Affiliation(s)
- Michelle L Giles
- Department of Infectious Diseases, University of Melbourne, Melbourne, Victoria, Australia
- Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Katie L Flanagan
- Department of Medicine, Launceston General Hospital, Launceston, Tasmania, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, Tasmania, Australia
- School of Medicine, University of Tasmania, Launceston, Tasmania, Australia
- School of Health and Biomedical Science, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|