1
|
Binayke A, Zaheer A, Vishwakarma S, Sharma P, Dandotiya J, Raghavan S, Gosain M, Singh S, Chattopadhyay S, Kaushal J, Madan U, Kshetrapal P, Batra G, Wadhwa N, Pandey AK, Bhatnagar S, Garg PK, Awasthi A. Understanding the landscape of the SARS-CoV-2-specific T cells post-omicron surge. J Med Virol 2024; 96:e29877. [PMID: 39169721 DOI: 10.1002/jmv.29877] [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: 05/30/2024] [Revised: 07/24/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024]
Abstract
Emerging evidence shows increased humoral response post-omicron surge, but research on T cell responses is limited. This study investigated the durability, magnitude, and breadth of SARS-CoV-2-spike-specific T cell responses in 216 two-dose vaccinated individuals pre- and post-omicron surge. Post-surge samples showed enhanced T cell responses, indicating widespread asymptomatic exposure to omicron. Further analysis of 105 individuals with multiple exposures to SARS-CoV-2 through boosters or infections showed that post-omicron, two-dose vaccinated individuals had T cell responses comparable to those of COVID-19 convalescents or boosted individuals. Additionally, we report cross-reactive T cell responses against omicron sub-variants, including BA2.86, remained strong, with preserved frequencies of spike-specific stem-cell-like memory T cells. In silico prediction indicates that mutated epitopes of JN.1 and KP.2 retain over 95.6% of their HLA binding capability. Overall, our data suggests that T cell responses are sustained, enhanced, and cross-reactive against emerging SARS-CoV-2 variants following symptomatic or asymptomatic omicron infection.
Collapse
Affiliation(s)
- Akshay Binayke
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
- Jawaharlal Nehru University, New Delhi, India
| | - Aymaan Zaheer
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Siddhesh Vishwakarma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Priyanka Sharma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Jyotsna Dandotiya
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
| | | | - Mudita Gosain
- Translational Health Science and Technology Institute, Faridabad, India
| | - Savita Singh
- Translational Health Science and Technology Institute, Faridabad, India
| | | | - Jyotsana Kaushal
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Upasna Madan
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
| | | | - Gaurav Batra
- Translational Health Science and Technology Institute, Faridabad, India
| | - Nitya Wadhwa
- Translational Health Science and Technology Institute, Faridabad, India
| | | | | | - Pramod Kumar Garg
- Translational Health Science and Technology Institute, Faridabad, India
- All India Institute of Medical Science, New Delhi, India
| | - Amit Awasthi
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
2
|
Pitiriga VC, Papamentzelopoulou M, Konstantinakou KE, Vasileiou IV, Sakellariou KS, Spyrou NI, Tsakris A. Persistence of T-Cell Immunity Responses against SARS-CoV-2 for over 12 Months Post COVID-19 Infection in Unvaccinated Individuals with No Detectable IgG Antibodies. Vaccines (Basel) 2023; 11:1764. [PMID: 38140169 PMCID: PMC10747023 DOI: 10.3390/vaccines11121764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/09/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Immune response to SARS-CoV-2 is crucial for preventing reinfection or reducing disease severity. T-cells' long-term protection, elicited either by COVID-19 vaccines or natural infection, has been extensively studied thus far; however, it is still attracting considerable scientific interest. The aim of the present epidemiological study was to define the levels of T-cellular immunity response in a specific group of unvaccinated individuals from the general population with a prior confirmed COVID-19 infection and no measurable levels of IgG antibodies. METHODS We performed a retrospective descriptive analysis of data collected from the medical records of consecutive unvaccinated individuals recovered from COVID-19, who had proceeded to a large private medical center in the Attica region from September 2021 to September 2022 in order to be examined on their own initiative for SARS-CoV-2 T-cell immunity response. The analysis of T-cell responses was divided into three time periods post infection: Group A: up to 6 months; Group B: 6-12 months; Group C: >12 months. The SARS-CoV-2 T-cell response was estimated against spike (S) and nucleocapsid (N) structural proteins by performing the T-SPOT. COVID test methodology. SARS-CoV-2 IgG antibody levels were measured by the SARS-CoV-2 IgG II Quant assay (Abbott Diagnostics). RESULTS A total of 182 subjects were retrospectively included in the study, 85 females (46.7%) and 97 (53.3%) males, ranging from 19 to 91 years old (mean 50.84 ± 17.2 years). Among them, 59 (32.4%) had been infected within the previous 6 months from the examination date (Group A), 69 (37.9%) had been infected within a time period > 6 months and <1 year (Group B) and 54 (29.7%) had been infected within a time period longer than 1 year from the examination date (Group C). Among the three groups, a positive T-cell reaction against the S antigen was reported in 47/58 (81%) of Group A, 61/69 (88.4%) of Group B and 40/54 (74.1%) of Group C (chi square, p = 0.27). T-cell reaction against the N antigen was present in 45/58 (77.6%) of Group A, 61/69 (88.4%) of Group B and 36/54 (66.7%) of Group C (chi square, p = 0.02). The median Spot-Forming Cells (SFC) count for the S antigen was 18 (range from 0-160) in Group A, 19 (range from 0-130) in Group B and 17 (range from 0-160) in Group C (Kruskal-Wallis test, p = 0.11; pairwise comparisons: groups A-B, p = 0.95; groups A-C, p = 0.89; groups B-C, p = 0.11). The median SFCs count for the N antigen was 14.5 (ranging from 0 to 116) for Group A, 24 (ranging from 0-168) in Group B and 16 (ranging from 0-112) for Group C (Kruskal-Wallis test, p = 0.01; pairwise comparisons: groups A-B, p = 0.02; groups A-C, p = 0.97; groups B-C, p = 0.03). CONCLUSIONS Our data suggest that protective adaptive T-cellular immunity following natural infection by SARS-CoV-2 may persist for over 12 months, despite the undetectable humoral element.
Collapse
Affiliation(s)
- Vassiliki C. Pitiriga
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece;
| | - Myrto Papamentzelopoulou
- Molecular Biology Unit, 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Kanella E. Konstantinakou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Irene V. Vasileiou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Konstantina S. Sakellariou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Natalia I. Spyrou
- Bioiatriki Healthcare Group, Kifisias 132 and Papada Street, 11526 Athens, Greece; (K.E.K.); (I.V.V.); (K.S.S.); (N.I.S.)
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece;
| |
Collapse
|
3
|
Long J, Soni M, Muranski P, Miller MJ, Conry-Cantilena C, De Giorgi V. Case Report: Kinetics and durability of humoral and cellular response of SARS-CoV-2 messenger RNA vaccine in a lung and kidney transplant recipient. Front Immunol 2023; 14:1207638. [PMID: 37465681 PMCID: PMC10350526 DOI: 10.3389/fimmu.2023.1207638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/09/2023] [Indexed: 07/20/2023] Open
Abstract
We present a case report of a 63-year-old female health care worker who is 15 years status post double lung transplant and six years status post living related donor kidney transplant who is healthy on a chronic immunosuppression regimen including prednisone, mycophenolate, and tacrolimus who received the SARS-CoV-2 mRNA vaccine (Pfizer-BioNTech BNT162b2) primary series and had poor initial humoral response to the COVID-19 mRNA vaccine, then demonstrated a robust, sustained immune response against S1 and S2 antigens for over seven months after receiving the recommended vaccine doses, including booster dose, without developing COVID-19 or other serious adverse events. Her immune response to vaccination indicates effective formation of anti-spike T cell memory despite chronic immunosuppression. This case report provides a comprehensive characterization of her immune response to this SARS-CoV-2 vaccination series. As vaccine effectiveness data is updated, and as better understanding of immune response including hybrid immunity emerges, these findings may reassure that recipients of SOTs may be capable of durable immune responses to emerging variants of SARS-CoV-2.
Collapse
Affiliation(s)
- James Long
- Infectious Diseases Section, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Mithil Soni
- Columbia Center for Translational Immunology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Pawel Muranski
- Columbia Center for Translational Immunology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Maureen J. Miller
- Infectious Diseases Section, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Cathleen Conry-Cantilena
- Infectious Diseases Section, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Valeria De Giorgi
- Infectious Diseases Section, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| |
Collapse
|
4
|
Fernández-Moreno R, Valle-Arroyo J, Páez-Vega A, Salinas A, Cano A, Pérez AB, Torre-Cisneros J, Cantisán S. Memory SARS-CoV-2 T-cell response in convalescent COVID-19 patients with undetectable specific IgG antibodies: a comparative study. Front Immunol 2023; 14:1142918. [PMID: 37180143 PMCID: PMC10169638 DOI: 10.3389/fimmu.2023.1142918] [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: 01/12/2023] [Accepted: 04/07/2023] [Indexed: 05/15/2023] Open
Abstract
Background During the COVID-19 pandemic, a variable percentage of patients with SARS-CoV-2 infection failed to elicit humoral response. This study investigates whether patients with undetectable SARS-CoV-2 IgG are able to generate SARS-CoV-2 memory T cells with proliferative capacity upon stimulation. Methods This cross-sectional study was conducted with convalescent COVID-19 patients, diagnosed with a positive real-time PCR (RT-PCR) from nasal and pharyngeal swab specimens. COVID-19 patients were enrolled ≥3 months after the last PCR positive. Proliferative T-cell response after whole blood stimulation was assessed using the FASCIA assay. Results A total of 119 participants (86 PCR-confirmed COVID-19 patients and 33 healthy controls) were randomly filtered from an initial cohort. Of these 86 patients, 59 had detectable (seropositive) and 27 had undetectable (seronegative) SARS-CoV-2 IgG. Seropositive patients were subclassified as asymptomatic/mild or severe according to the oxygen supplementation requirement. SARS-CoV-2 CD3+ and CD4+ T cells showed significantly lower proliferative response in seronegative than in seropositive patients. The ROC curve analysis indicated that ≥ 5 CD4+ blasts/μL of blood defined a "positive SARS-CoV-2 T cell response". According to this cut-off, 93.2% of seropositive patients had a positive T-cell response compared to 50% of seronegative patients and 20% of negative controls (chi-square; p < 0.001). Conclusions This proliferative assay is useful not only to discriminate convalescent patients from negative controls, but also to distinguish seropositive patients from those with undetectable SARS-CoV-2 IgG antibodies. Memory T cells in seronegative patients are able to respond to SARSCoV-2 peptides, although at a lower magnitude than seropositive patients.
Collapse
Affiliation(s)
- Raquel Fernández-Moreno
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
| | - Jorge Valle-Arroyo
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
| | - Aurora Páez-Vega
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
| | - Ana Salinas
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
| | - Angela Cano
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
- Infectious Diseases Unit, Reina Sofía University Hospital, Cordoba, Spain
| | - Ana B Pérez
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
- Microbiology Unit, Reina Sofía University Hospital, Cordoba, Spain
| | - Julián Torre-Cisneros
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
- Infectious Diseases Unit, Reina Sofía University Hospital, Cordoba, Spain
| | - Sara Cantisán
- Spanish Network for Research in Infectious Diseases (REIPI), Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases (GC-03) and Clinical and Molecular Microbiology (GC-24) Groups, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
- Infectious Diseases Unit, Reina Sofía University Hospital, Cordoba, Spain
| |
Collapse
|
5
|
Løken RØ, Fevang B. Cellular immunity in COVID-19 and other infections in Common variable immunodeficiency. Front Immunol 2023; 14:1124279. [PMID: 37180118 PMCID: PMC10173090 DOI: 10.3389/fimmu.2023.1124279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
COVID-19 has shed light on the role of cellular immunity in the absence of humoral response in different patient groups. Common variable immunodeficiency (CVID) is characterized by impaired humoral immunity but also an underlying T-cell dysregulation. The impact of T-cell dysregulation on cellular immunity in CVID is not clear, and this review summarizes available literature on cellular immunity in CVID with a particular focus on COVID-19. Overall mortality of COVID-19 in CVID is difficult to assess, but seems not significantly elevated, and risk factors for severe disease mirrors that of the general population, including lymphopenia. Most CVID patients have a significant T-cell response to COVID-19 disease with possible cross-reactivity to endemic coronaviruses. Several studies find a significant but impaired cellular response to basal COVID-19 mRNA vaccination that is independent of an antibody response. CVID patients with infection only have better cellular responses to vaccine in one study, but there is no clear association to T-cell dysregulation. Cellular response wane over time but responds to a third booster dose of vaccine. Opportunistic infection as a sign of impaired cellular immunity in CVID is rare but is related to the definition of the disease. CVID patients have a cellular response to influenza vaccine that in most studies is comparable to healthy controls, and annual vaccination against seasonal influenza should be recommended. More research is required to clarify the effect of vaccines in CVID with the most immediate issue being when to booster the COVID-19 vaccine.
Collapse
Affiliation(s)
- Ragnhild Øye Løken
- Section of Clinical Immunology and Infectious Diseases, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital, Oslo, Norway
| | - Børre Fevang
- Section of Clinical Immunology and Infectious Diseases, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital, Oslo, Norway
- Centre for Rare Disorders, Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- *Correspondence: Børre Fevang,
| |
Collapse
|
6
|
Kombe Kombe AJ, Biteghe FAN, Ndoutoume ZN, Jin T. CD8 + T-cell immune escape by SARS-CoV-2 variants of concern. Front Immunol 2022; 13:962079. [PMID: 36389664 PMCID: PMC9647062 DOI: 10.3389/fimmu.2022.962079] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/03/2022] [Indexed: 07/30/2023] Open
Abstract
Despite the efficacy of antiviral drug repositioning, convalescent plasma (CP), and the currently available vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the worldwide coronavirus disease 2019 (COVID-19) pandemic is still challenging because of the ongoing emergence of certain new SARS-CoV-2 strains known as variants of concern (VOCs). Mutations occurring within the viral genome, characterized by these new emerging VOCs, confer on them the ability to efficiently resist and escape natural and vaccine-induced humoral and cellular immune responses. Consequently, these VOCs have enhanced infectivity, increasing their stable spread in a given population with an important fatality rate. While the humoral immune escape process is well documented, the evasion mechanisms of VOCs from cellular immunity are not well elaborated. In this review, we discussed how SARS-CoV-2 VOCs adapt inside host cells and escape anti-COVID-19 cellular immunity, focusing on the effect of specific SARS-CoV-2 mutations in hampering the activation of CD8+ T-cell immunity.
Collapse
Affiliation(s)
- Arnaud John Kombe Kombe
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | | | - Zélia Nelly Ndoutoume
- The Second Clinical School, Medical Imaging, Chongqing Medical University, Chongqing, China
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Laboratory of Structural Immunology, Chinese Academic of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Chinese Academic of Sciences (CAS) Center for Excellence in Molecular Cell Science, Chinese Academy of Science, Shanghai, China
| |
Collapse
|
7
|
Murphy RL, Paramithiotis E, Sugden S, Chermak T, Lambert B, Montamat-Sicotte D, Mattison J, Steinhubl S. The need for more holistic immune profiling in next-generation SARS-CoV-2 vaccine trials. Front Immunol 2022; 13:923106. [PMID: 36211354 PMCID: PMC9533322 DOI: 10.3389/fimmu.2022.923106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/26/2022] [Indexed: 11/14/2022] Open
Abstract
First-generation anit-SARS-CoV-2 vaccines were highly successful. They rapidly met an unforeseen emergency need, saved millions of lives, and simultaneously eased the burden on healthcare systems worldwide. The first-generation vaccines, however, focused too narrowly on antibody-based immunity as the sole marker of vaccine trial success, resulting in large knowledge gaps about waning vaccine protection, lack of vaccine robustness to viral mutation, and lack of efficacy in immunocompromised populations. Detailed reviews of first-generation vaccines, including their mode of action and geographical distribution, have been published elsewhere. Second-generation clinical trials must address these gaps by evaluating a broader range of immune markers, including those representing cell-mediated immunity, to ensure the most protective and long-lasting vaccines are brought to market.
Collapse
Affiliation(s)
- Robert L. Murphy
- Northwestern University, Evanston, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- *Correspondence: Robert L. Murphy,
| | | | | | | | - Bruce Lambert
- Northwestern University, Evanston, IL, United States
| | | | | | | |
Collapse
|
8
|
Maamari KA, Busaidi IA, Kindi MA, Zadjali F, BaAlawi F, Anesta W, Amri KA, Albalushi W, Balushi HA, Amri AA, Aljufaili M, Al-Busaidi M, Muharrmi ZA, Balkhair A, Riyami NA, Ghanim Z, Alshekaili J. Short and long-term immune changes in different severity groups of COVID-19 disease. Int J Infect Dis 2022; 122:776-784. [PMID: 35840099 PMCID: PMC9284586 DOI: 10.1016/j.ijid.2022.07.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND There are limited data on short- versus long-term changes in adaptive immune response across different COVID-19 disease severity groups. METHODS A multicenter prospective study of 140 adult patients with COVID-19 (a total of 325 samples) were analyzed for inflammatory markers and lymphocyte subsets at presentation, week 2, and week 24. RESULTS Inflammatory markers at presentation were higher in the critical/severe than in moderate and mild groups. A predominance of memory B cell response in the mild and moderate group was noted by week 2. In contrast, the immune system in the severe/critical group was dysfunctional, with expansion of exhausted CD8+ T cells and atypical memory B cells. By 24 weeks, there was a possible trend of normalization. CONCLUSION There was substantial difference in the degree of inflammation and distribution of different B and T cell subsets in the different disease severity groups. Despite the initial dysfunctional immune response in the severe/critical group, a comparable memory B and CD8+ T cell responses to the mild group was achieved at 24 weeks.
Collapse
Affiliation(s)
- Khuloud Al Maamari
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Ibrahim Al Busaidi
- Department of Medicine, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Mahmood Al Kindi
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Fahad Zadjali
- Department of Clinical Biochemistry, Sultan Qaboos University, Sultanate of Oman
| | - Fatma BaAlawi
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Wijesinghe Anesta
- Department of internal medicine, Armed Forces Hospital, Sultanate of Oman
| | - Kawthar Al Amri
- Department of internal medicine, Armed Forces Hospital, Sultanate of Oman
| | - Wafa Albalushi
- Department of Nursing, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Hamed Al Balushi
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Ayman Al Amri
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Mahmood Aljufaili
- Department of Emergency, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Mujahid Al-Busaidi
- Department of Medicine, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Zakariya Al Muharrmi
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Abdullah Balkhair
- Department of Medicine, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Nafila Al Riyami
- Department of Clinical Biochemistry, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Zahraa Ghanim
- Department of Anaesthesia, Sultan Qaboos University Hospital, Sultanate of Oman
| | - Jalila Alshekaili
- Department of Microbiology and Immunology, Sultan Qaboos University Hospital, Sultanate of Oman,Corresponding author
| |
Collapse
|
9
|
Shrestha LB, Foster C, Rawlinson W, Tedla N, Bull RA. Evolution of the SARS-CoV-2 omicron variants BA.1 to BA.5: Implications for immune escape and transmission. Rev Med Virol 2022; 32:e2381. [PMID: 35856385 PMCID: PMC9349777 DOI: 10.1002/rmv.2381] [Citation(s) in RCA: 276] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 12/15/2022]
Abstract
The first dominant SARS-CoV-2 Omicron variant BA.1 harbours 35 mutations in its Spike protein from the original SARS-CoV-2 variant that emerged late 2019. Soon after its discovery, BA.1 rapidly emerged to become the dominant variant worldwide and has since evolved into several variants. Omicron is of major public health concern owing to its high infectivity and antibody evasion. This review article examines the theories that have been proposed on the evolution of Omicron including zoonotic spillage, infection in immunocompromised individuals and cryptic spread in the community without being diagnosed. Added to the complexity of Omicron's evolution are the multiple reports of recombination events occurring between co-circulating variants of Omicron with Delta and other variants such as XE. Current literature suggests that the combination of the novel mutations in Omicron has resulted in the variant having higher infectivity than the original Wuhan-Hu-1 and Delta variant. However, severity is believed to be less owing to the reduced syncytia formation and lower multiplication in the human lung tissue. Perhaps most challenging is that several studies indicate that the efficacy of the available vaccines have been reduced against Omicron variant (8-127 times reduction) as compared to the Wuhan-Hu-1 variant. The administration of booster vaccine, however, compensates with the reduction and improves the efficacy by 12-35 fold. Concerningly though, the broadly neutralising monoclonal antibodies, including those approved by FDA for therapeutic use against previous SARS-CoV-2 variants, are mostly ineffective against Omicron with the exception of Sotrovimab and recent reports suggest that the Omicron BA.2 is also resistant to Sotrovimab. Currently two new Omicron variants BA.4 and BA.5 are emerging and are reported to be more transmissible and resistant to immunity generated by previous variants including Omicron BA.1 and most monoclonal antibodies. As new variants of SARS-CoV-2 will likely continue to emerge it is important that the evolution, and biological consequences of new mutations, in existing variants be well understood.
Collapse
Affiliation(s)
- Lok Bahadur Shrestha
- School of Medical SciencesFaculty of MedicineUNSWSydneyNew South WalesAustralia
- The Kirby InstituteUNSWSydneyNew South WalesAustralia
| | - Charles Foster
- School of Medical SciencesFaculty of MedicineUNSWSydneyNew South WalesAustralia
- Serology and Virology DivisionDepartment of MicrobiologyNew South Wales Health PathologySydneyNew South WalesAustralia
| | - William Rawlinson
- School of Medical SciencesFaculty of MedicineUNSWSydneyNew South WalesAustralia
- Serology and Virology DivisionDepartment of MicrobiologyNew South Wales Health PathologySydneyNew South WalesAustralia
| | - Nicodemus Tedla
- School of Medical SciencesFaculty of MedicineUNSWSydneyNew South WalesAustralia
| | - Rowena A. Bull
- School of Medical SciencesFaculty of MedicineUNSWSydneyNew South WalesAustralia
- The Kirby InstituteUNSWSydneyNew South WalesAustralia
| |
Collapse
|
10
|
Chiuppesi F, Zaia JA, Faircloth K, Johnson D, Ly M, Karpinski V, La Rosa C, Drake J, Marcia J, Acosta AM, Dempsey S, Taplitz RA, Zhou Q, Park Y, Ortega Francisco S, Kaltcheva T, Frankel PH, Rosen S, Wussow F, Dadwal S, Diamond DJ. Vaccine-induced spike- and nucleocapsid-specific cellular responses maintain potent cross-reactivity to SARS-CoV-2 Delta and Omicron variants. iScience 2022; 25:104745. [PMID: 35846380 PMCID: PMC9272674 DOI: 10.1016/j.isci.2022.104745] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/16/2022] [Accepted: 07/06/2022] [Indexed: 01/06/2023] Open
Abstract
Cell-mediated immunity may contribute to providing protection against SARS-CoV-2 and its variants of concern (VOC). We developed COH04S1, a synthetic multiantigen modified vaccinia Ankara (MVA)-based COVID-19 vaccine that stimulated potent spike (S) and nucleocapsid (N) antigen-specific humoral and cellular immunity in a phase 1 clinical trial in healthy adults. Here, we show that individuals vaccinated with COH04S1 or mRNA vaccine BNT162b2 maintain robust cross-reactive cellular immunity for six or more months post-vaccination. Although neutralizing antibodies induced in COH04S1- and BNT162b2-vaccinees showed reduced activity against Delta and Omicron variants compared to ancestral SARS-CoV-2, S-specific T cells elicited in both COH04S1- and BNT162b2-vaccinees and N-specific T cells elicited in COH04S1-vaccinees demonstrated potent and equivalent cross-reactivity against ancestral SARS-CoV-2 and the major VOC. These results suggest that vaccine-induced T cells to S and N antigens may constitute a critical second line of defense to provide long-term protection against SARS-CoV-2 VOC.
Collapse
Affiliation(s)
- Flavia Chiuppesi
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - John A. Zaia
- Center for Gene Therapy, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Katelyn Faircloth
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Daisy Johnson
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Minh Ly
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Veronica Karpinski
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Corinna La Rosa
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Jennifer Drake
- Clinical Trials Office, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Joan Marcia
- Clinical Trials Office, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Ann Marie Acosta
- Clinical Trials Office, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Shannon Dempsey
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Randy A. Taplitz
- Division of Infectious Diseases, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
- Department of Medicine, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Qiao Zhou
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Yoonsuh Park
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Sandra Ortega Francisco
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Teodora Kaltcheva
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Paul H. Frankel
- Department of Biostatistics, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Steven Rosen
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Felix Wussow
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Sanjeet Dadwal
- Division of Infectious Diseases, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
- Department of Medicine, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA 91010, USA
| | - Don J. Diamond
- Department of Hematology and HCT and Hematologic Malignancies Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA
- Corresponding author
| |
Collapse
|
11
|
Schwarz T, Otto C, Jones TC, Pache F, Schindler P, Niederschweiberer M, Schmidt FA, Drosten C, Corman VM, Ruprecht K. Preserved T cell responses to SARS-CoV-2 in anti-CD20 treated multiple sclerosis. Mult Scler 2022; 28:1041-1050. [PMID: 35575234 PMCID: PMC9131414 DOI: 10.1177/13524585221094478] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background: Optimal management of anti-CD20-treated patients with multiple sclerosis (pwMS) is an important clinical task during the current severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic. Objectives: To characterize humoral and cellular immune responses to SARS-CoV-2 vaccinations/infections in a longitudinal cohort of anti-CD20 treated (n = 175) and anti-CD20 therapy-naïve (n = 41) pwMS. Methods: Anti-SARS-CoV-2 spike protein immunoglobulin G (IgG) and IgA, virus neutralizing capacity, IgG avidity and SARS-CoV-2-specific T cells were determined. Results: Following two SARS-CoV-2 vaccinations, not only SARS-CoV-2 spike protein IgG and IgA, but also neutralizing capacity and avidity of SARS-CoV-2 IgG were lower in anti-CD20-treated (n = 51) than in anti-CD20 therapy-naïve pwMS (n = 14) and in healthy controls (HC, n = 19). However, in all anti-CD20-treated pwMS vaccinated twice (n = 26) or infected with SARS-CoV-2 (n = 2), in whom SARS-CoV-2-specific T cells were measured, SARS-CoV-2-specific T cells were detectable, at levels similar to those of twice-vaccinated anti-CD20 therapy-naïve pwMS (n = 7) and HC (n = 19). SARS-CoV-2-S1 IgG levels (r = 0.42, p = 0.002), antibody avidity (r = 0.7, p < 0.001), and neutralizing capacity (r = 0.44, p = 0.03) increased with time between anti-CD20 infusion and second vaccination. Based on detection of SARS-CoV-2 antibodies, SARS-CoV-2 infections occurred in 4 out of 175 (2.3%) anti-CD20-treated pwMS, all of whom recovered fully. Conclusions: These findings should inform treatment decisions and SARS-CoV-2 vaccination management in pwMS.
Collapse
Affiliation(s)
- Tatjana Schwarz
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany/German Centre for Infection Research (DZIF), Berlin, Germany
| | - Carolin Otto
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Terry C Jones
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany/German Centre for Infection Research (DZIF), Berlin, Germany
| | - Florence Pache
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Patrick Schindler
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Moritz Niederschweiberer
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Felix A Schmidt
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany/German Centre for Infection Research (DZIF), Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité -Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany/German Center for Infection Research (DZIF), Berlin, Germany/Labor Berlin-Charité Vivantes GmbH, Berlin, Germany
| | - Klemens Ruprecht
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
12
|
Kilpeläinen A, Jimenez-Moyano E, Blanch-Lombarte O, Ouchi D, Peña R, Quirant-Sanchez B, Perez-Caballero R, Chamorro A, Blanco I, Martínez-Caceres E, Paredes R, Mateu L, Carrillo J, Blanco J, Brander C, Massanella M, Clotet B, Prado JG. Skewed Cellular Distribution and Low Activation of Functional T-Cell Responses in SARS-CoV-2 Non-Seroconvertors. Front Immunol 2022; 13:815041. [PMID: 35619701 PMCID: PMC9128381 DOI: 10.3389/fimmu.2022.815041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 03/29/2022] [Indexed: 12/21/2022] Open
Abstract
The role of T cells in the control of SARS-CoV-2 infection has been underestimated in favor of neutralizing antibodies. However, cellular immunity is essential for long-term viral control and protection from disease severity. To understand T-cell immunity in the absence of antibody generation we focused on a group of SARS-CoV-2 Non-Seroconvertors (NSC) recovered from infection. We performed an immune comparative analysis of SARS-CoV-2 infected individuals stratified by the absence or presence of seroconversion and disease severity. We report high levels of total naïve and low effector CD8+ T cells in NSC. Moreover, reduced levels of T-cell activation monitored by PD-1 and activation-induced markers were observed in the context of functional SARS-CoV-2 T-cell responses. Longitudinal data indicate the stability of the NSC phenotype over three months of follow-up after infection. Together, these data characterized distinctive immunological traits in NSC including skewed cellular distribution, low activation and functional SARS-CoV-2 T-cell responses. This data highlights the value of T-cell immune monitoring in populations with low seroconversion rates in response to SARS-CoV-2 infection and vaccination.
Collapse
Affiliation(s)
- Athina Kilpeläinen
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | | | | | - Dan Ouchi
- IrsiCaixa AIDS Research Institute, Badalona, Spain
| | - Ruth Peña
- IrsiCaixa AIDS Research Institute, Badalona, Spain
| | - Bibiana Quirant-Sanchez
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Cell Biology, Physiology, Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Immunology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | - Anna Chamorro
- Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Ignacio Blanco
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Clinical Genetics and Genetic Counseling Program, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Eva Martínez-Caceres
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Cell Biology, Physiology, Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Immunology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Roger Paredes
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
| | - Lourdes Mateu
- Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- CIBERINFEC, ISCIII, Madrid, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- CIBERINFEC, ISCIII, Madrid, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- CIBERINFEC, ISCIII, Madrid, Spain
- ICREA, Barcelona, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
| | - Marta Massanella
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- CIBERINFEC, ISCIII, Madrid, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Lluita contra la SIDA Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Catalonia, Spain
| | - Julia G. Prado
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- CIBERINFEC, ISCIII, Madrid, Spain
| |
Collapse
|
13
|
Wirsching S, Harder L, Heymanns M, Gröndahl B, Hilbert K, Kowalzik F, Meyer C, Gehring S. Long-Term, CD4 + Memory T Cell Response to SARS-CoV-2. Front Immunol 2022; 13:800070. [PMID: 35514974 PMCID: PMC9065554 DOI: 10.3389/fimmu.2022.800070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/28/2022] [Indexed: 01/08/2023] Open
Abstract
The first cases of coronavirus disease-19 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were reported by Chinese authorities at the end of 2019. The disease spread quickly and was declared a global pandemic shortly thereafter. To respond effectively to infection and prevent viral spread, it is important to delineate the factors that affect protective immunity. Herein, a cohort of convalescent healthcare workers was recruited and their immune responses were studied over a period of 3 to 9 months following the onset of symptoms. A cross-reactive T cell response to SARS-CoV-2 and endemic coronaviruses, i.e., OC43 and NL63, was demonstrated in the infected, convalescent cohort, as well as a cohort composed of unexposed individuals. The convalescent cohort, however, displayed an increased number of SARS-CoV-2-specific CD4+ T cells relative to the unexposed group. Moreover, unlike humoral immunity and quickly decreasing antibody titers, T cell immunity in convalescent individuals was maintained and stable throughout the study period. This study also suggests that, based on the higher CD4 T cell memory response against nucleocapsid antigen, future vaccine designs may include nucleocapsid as an additional antigen along with the spike protein.
Collapse
Affiliation(s)
- Sebastian Wirsching
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Laura Harder
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Markus Heymanns
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Britta Gröndahl
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Katja Hilbert
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Frank Kowalzik
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Claudius Meyer
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stephan Gehring
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
14
|
Odak I, Schultze-Florey CR, Hammerschmidt SI, Ritter C, Willenzon S, Friedrichsen M, Ravens I, Sikora R, Bayir LM, Gutierrez Jauregui R, Bernhardt G, Stankov MV, Cossmann A, Hansen G, Krey T, Cornberg M, Koenecke C, Behrens GMN, Bošnjak B, Förster R. Longitudinal Tracking of Immune Responses in COVID-19 Convalescents Reveals Absence of Neutralization Activity Against Omicron and Staggered Impairment to Other SARS-CoV-2 Variants of Concern. Front Immunol 2022; 13:863039. [PMID: 35359969 PMCID: PMC8964088 DOI: 10.3389/fimmu.2022.863039] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Evaluating long-term protection against SARS-CoV-2 variants of concern in convalescing individuals is of high clinical relevance. In this prospective study of a cohort of 46 SARS-CoV-2 patients infected with the Wuhan strain of SARS-CoV-2 we longitudinally analyzed changes in humoral and cellular immunity upon early and late convalescence. Antibody neutralization capacity was measured by surrogate virus neutralization test and cellular responses were investigated with 31-colour spectral flow cytometry. Spike-specific, isotype-switched B cells developed already during the disease phase, showed a memory phenotype and did not decrease in numbers even during late convalescence. Otherwise, no long-lasting perturbations of the immune compartment following COVID-19 clearance were observed. During convalescence anti-Spike (S1) IgG antibodies strongly decreased in all patients. We detected neutralizing antibodies against the Wuhan strain as well as the Alpha and Delta but not against the Beta, Gamma or Omicron variants for up to 7 months post COVID-19. Furthermore, correlation analysis revealed a strong association between sera anti-S1 IgG titers and their neutralization capacity against the Wuhan strain as well as Alpha and Delta. Overall, our data suggest that even 7 month after the clearance of COVID-19 many patients possess a protective layer of immunity, indicated by the persistence of Spike-specific memory B cells and by the presence of neutralizing antibodies against the Alpha and Delta variants. However, lack of neutralizing antibodies against the Beta, Gamma and Omicron variants even during the peak response is of major concern as this indicates viral evasion of the humoral immune response.
Collapse
Affiliation(s)
- Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christian R. Schultze-Florey
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | | | | | | | | - Inga Ravens
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Ruth Sikora
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Lâle M. Bayir
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Günter Bernhardt
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Metodi V. Stankov
- Clinic Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Anne Cossmann
- Clinic Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Guido Hansen
- Institute of Biochemistry, University of Lübeck, Lübeck, Germany
| | - Thomas Krey
- Institute of Biochemistry, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg-Lübeck-Borstel-Riems, Brunswick, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Markus Cornberg
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Sites Hannover-Braunschweig, Brunswick, Germany
- Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Christian Koenecke
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Georg M. N. Behrens
- Clinic Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Sites Hannover-Braunschweig, Brunswick, Germany
- Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
| | - Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Sites Hannover-Braunschweig, Brunswick, Germany
| |
Collapse
|
15
|
Steiner S, Schwarz T, Corman VM, Gebert L, Kleinschmidt MC, Wald A, Gläser S, Kruse JM, Zickler D, Peric A, Meisel C, Meyer T, Staudacher OL, Wittke K, Kedor C, Bauer S, Besher NA, Kalus U, Pruß A, Drosten C, Volk HD, Scheibenbogen C, Hanitsch LG. SARS-CoV-2 T Cell Response in Severe and Fatal COVID-19 in Primary Antibody Deficiency Patients Without Specific Humoral Immunity. Front Immunol 2022; 13:840126. [PMID: 35359967 PMCID: PMC8960624 DOI: 10.3389/fimmu.2022.840126] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 12/19/2022] Open
Abstract
Morbidity and mortality of COVID-19 is increased in patients with inborn errors of immunity (IEI). Age and comorbidities and also impaired type I interferon immunity were identified as relevant risk factors. In patients with primary antibody deficiency (PAD) and lack of specific humoral immune response to SARS-CoV-2, clinical disease outcome is very heterogeneous. Despite extensive clinical reports, underlying immunological mechanisms are poorly characterized and levels of T cellular and innate immunity in severe cases remain to be determined. In the present study, we report clinical and immunological findings of 5 PAD patients with severe and fatal COVID-19 and undetectable specific humoral immune response to SARS-CoV-2. Reactive T cells to SARS-CoV-2 spike (S) and nucleocapsid (NCAP) peptide pools were analyzed comparatively by flow cytometry in PAD patients, convalescents and naïve healthy individuals. All examined PAD patients developed a robust T cell response. The presence of polyfunctional cytokine producing activated CD4+ T cells indicates a memory-like phenotype. An analysis of innate immune response revealed elevated CD169 (SIGLEC1) expression on monocytes, a surrogate marker for type I interferon response, and presence of type I interferon autoantibodies was excluded. SARS-CoV-2 RNA was detectable in peripheral blood in three severe COVID-19 patients with PAD. Viral clearance in blood was observed after treatment with COVID-19 convalescent plasma/monoclonal antibody administration. However, prolonged mucosal viral shedding was observed in all patients (median 67 days) with maximum duration of 127 days. PAD patients without specific humoral SARS-CoV-2 immunity may suffer from severe or fatal COVID-19 despite robust T cell and normal innate immune response. Intensified monitoring for long persistence of SARS-CoV-2 viral shedding and (prophylactic) convalescent plasma/specific IgG as beneficial treatment option in severe cases with RNAemia should be considered in seronegative PAD patients.
Collapse
Affiliation(s)
- Sophie Steiner
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1 and Berlin Institute of Health, Berlin, Germany
| | - Tatjana Schwarz
- Institute of Virology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and German Centre for Infection Research, Associated Partner, Charitéplatz 1, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Victor M. Corman
- Institute of Virology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and German Centre for Infection Research, Associated Partner, Charitéplatz 1, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Laura Gebert
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1 and Berlin Institute of Health, Berlin, Germany
| | - Malte C. Kleinschmidt
- Department of Infectious Diseases and Respiratory Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexandra Wald
- Department of Pulmonary Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Sven Gläser
- Department of Pulmonary Medicine and Infectious Diseases, Vivantes-Klinikum Neukölln, Berlin, Germany
| | - Jan M. Kruse
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Daniel Zickler
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alexander Peric
- Department of Pulmonary Medicine and Infectious Diseases, Vivantes-Klinikum Friedrichshain, Berlin, Germany
| | - Christian Meisel
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1 and Berlin Institute of Health, Berlin, Germany
- Department of Immunology, Labor Berlin GmbH, Berlin, Germany
| | - Tim Meyer
- Department of Immunology, Labor Berlin GmbH, Berlin, Germany
| | - Olga L. Staudacher
- Department of Immunology, Labor Berlin GmbH, Berlin, Germany
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kirsten Wittke
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1 and Berlin Institute of Health, Berlin, Germany
| | - Claudia Kedor
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1 and Berlin Institute of Health, Berlin, Germany
| | - Sandra Bauer
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1 and Berlin Institute of Health, Berlin, Germany
| | - Nabeel Al Besher
- Institute of Transfusion Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrich Kalus
- Institute of Transfusion Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Axel Pruß
- Institute of Transfusion Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and German Centre for Infection Research, Associated Partner, Charitéplatz 1, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Hans-Dieter Volk
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1 and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Charitéplatz 1, Berlin, Germany
- Berlin Center for Advanced Therapies, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Scheibenbogen
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1 and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Charitéplatz 1, Berlin, Germany
| | - Leif G. Hanitsch
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1 and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
16
|
Kato H, Miyakawa K, Ohtake N, Yamaoka Y, Yajima S, Yamazaki E, Shimada T, Goto A, Nakajima H, Ryo A. Vaccine-induced humoral response against SARS-CoV-2 dramatically declined but cellular immunity possibly remained at 6 months post BNT162b2 vaccination. Vaccine 2022; 40:2652-2655. [PMID: 35370020 PMCID: PMC8960126 DOI: 10.1016/j.vaccine.2022.03.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023]
Abstract
To evaluate vaccine-induced humoral and cell-mediated immunity at 6 months after completion of two doses of BNT162b2 vaccination, immunoglobulin G against SARS-CoV-2 spike protein (SP IgG), 50% neutralizing antibody (NT50), and spot-forming cell (SFC) counts were evaluated by interferon-γ releasing ELISpot assay of 98 healthy subjects (median age, 43 years). The geometric mean titers of SP IgG and NT50 decreased from 95.2 (95% confidence interval (CI) 79.8–113.4) to 5.7 (95% CI 4.9–6.7) and from 680.4 (588.0–787.2) to 130.4 (95% CI 104.2–163.1), respectively, at 3 weeks and 6 months after the vaccination. SP IgG titer was negatively correlated with age and alcohol consumption. Spot-forming cell counts at 6 months did not correlate with age, gender, and other parameters of the patients. SP IgG, NT50, and SFC titers were elevated in the breakthrough infected subjects. Although the levels of vaccine-induced antibodies dramatically declined at 6 months after vaccination, a certain degree of cellular immunity was observed irrespective of the age.
Collapse
Affiliation(s)
- Hideaki Kato
- Infection Prevention and Control Department, Yokohama City University Hospital, Yokohama, Japan; Department of Hematology and Clinical Immunology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Kei Miyakawa
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Norihisa Ohtake
- Bioscience Division, Research and Development Department, Tosoh Corporation, Tokyo Research Center, Kanagawa, Japan; Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yutaro Yamaoka
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan; Life Science Laboratory, Technology and Development Division, Kanto Chemical Co, Inc., Isehara, Japan
| | - Satoshi Yajima
- Clinical Laboratory Department, Yokohama City University Hospital, Yokohama, Japan
| | - Etsuko Yamazaki
- Clinical Laboratory Department, Yokohama City University Hospital, Yokohama, Japan
| | - Tomoko Shimada
- Nursing Department, Yokohama City University Hospital, Yokohama, Japan
| | - Atsushi Goto
- Department of Health Data Science, Yokohama City University Graduate School of Data Science, Yokohama, Japan
| | - Hideaki Nakajima
- Department of Hematology and Clinical Immunology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan.
| |
Collapse
|
17
|
Usai C, Gibbons JM, Pade C, Li W, Jacobs SRM, McKnight Á, Kennedy PTF, Gill US. The β-NGF/TrkA Signalling Pathway Is Associated With the Production of Anti-Nucleoprotein IgG in Convalescent COVID-19. Front Immunol 2022; 12:813300. [PMID: 35095908 PMCID: PMC8795736 DOI: 10.3389/fimmu.2021.813300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/16/2021] [Indexed: 01/10/2023] Open
Abstract
Background The presentation of SARS-CoV-2 infection varies from asymptomatic to severe COVID-19. Similarly, high variability in the presence, titre and duration of specific antibodies has been reported. While some host factors determining these differences, such as age and ethnicity have been identified, the underlying molecular mechanisms underpinning these differences remain poorly defined. Methods We analysed serum and PBMC from 17 subjects with a previous PCR-confirmed SARS-CoV-2 infection and 10 unexposed volunteers following the first wave of the pandemic, in the UK. Anti-NP IgG and neutralising antibodies were measured, as well as a panel of infection and inflammation related cytokines. The virus-specific T cell response was determined by IFN-γ ELISPOT and flow cytometry after overnight incubation of PBMCs with pools of selected SARS-CoV-2 specific peptides. Results Seven of 17 convalescent subjects had undetectable levels of anti-NP IgG, and a positive correlation was shown between anti-NP IgG levels and the titre of neutralising antibodies (IC50). In contrast, a discrepancy was noted between antibody levels and T cell IFN-γ production by ELISpot following stimulation with specific peptides. Among the analysed cytokines, β-NGF and IL-1α levels were significantly different between anti-NP positive and negative subjects, and only β-NGF significantly correlated with anti-NP positivity. Interestingly, CD4+ T cells of anti-NP negative subjects expressed lower amounts of the β-NGF-specific receptor TrkA. Conclusions Our results suggest that the β-NGF/TrkA signalling pathway is associated with the production of anti-NP specific antibody in mild SARS-CoV-2 infection and the mechanistic regulation of this pathway in COVID-19 requires further investigation.
Collapse
Affiliation(s)
- Carla Usai
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Joseph M. Gibbons
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Corinna Pade
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Wenhao Li
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Health National Health Service (NHS) Trust, The Royal London Hospital, London, United Kingdom
| | - Sabina R. M. Jacobs
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Áine McKnight
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Patrick T. F. Kennedy
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Health National Health Service (NHS) Trust, The Royal London Hospital, London, United Kingdom
| | - Upkar S. Gill
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Health National Health Service (NHS) Trust, The Royal London Hospital, London, United Kingdom
| |
Collapse
|
18
|
Awuah A, Zamani A, Tahami F, Davis M, Grandjean L, Buckland M, Gilmour K. T cell responses to SARS-CoV-2 in healthy controls and primary immunodeficiency patients. Clin Exp Immunol 2022; 207:uxac001. [PMID: 35020892 PMCID: PMC8807284 DOI: 10.1093/cei/uxac001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/03/2021] [Accepted: 01/05/2022] [Indexed: 01/03/2023] Open
Abstract
Understanding the T cell response to SARS-CoV-2 is key in patients who lack antibody production. We demonstrate the applicability of a functional assay to measure the T cell response in a cohort of patients with immunodeficiency.
Collapse
Affiliation(s)
- Arnold Awuah
- Department of Immunology, Great Ormond Street Hospital (GOSH), London, UK
| | - Ava Zamani
- Affiliate of Department of Immunology, Great Ormond Street Hospital (GOSH), London, UK
| | - Fariba Tahami
- Department of Immunology, Great Ormond Street Hospital (GOSH), London, UK
| | - Mark Davis
- Department of Immunology, Great Ormond Street Hospital (GOSH), London, UK
| | - Louis Grandjean
- Department of Infectious Diseases, Great Ormond Street Hospital (GOSH), London, UK
| | - Matthew Buckland
- Department of Immunology, Great Ormond Street Hospital (GOSH), London, UK
| | - Kimberly Gilmour
- Department of Immunology, Great Ormond Street Hospital (GOSH), London, UK
| |
Collapse
|
19
|
Ahmed SF, Quadeer AA, McKay MR. SARS-CoV-2 T Cell Responses Elicited by COVID-19 Vaccines or Infection Are Expected to Remain Robust against Omicron. Viruses 2022; 14:79. [PMID: 35062283 PMCID: PMC8781795 DOI: 10.3390/v14010079] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/26/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023] Open
Abstract
Omicron, the most recent SARS-CoV-2 variant of concern (VOC), harbours multiple mutations in the spike protein that were not observed in previous VOCs. Initial studies suggest Omicron to substantially reduce the neutralizing capability of antibodies induced from vaccines and previous infection. However, its effect on T cell responses remains to be determined. Here, we assess the effect of Omicron mutations on known T cell epitopes and report data suggesting T cell responses to remain broadly robust against this new variant.
Collapse
Affiliation(s)
- Syed Faraz Ahmed
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China;
| | - Ahmed Abdul Quadeer
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China;
| | - Matthew R. McKay
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China;
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China
- Department of Electrical and Electronic Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| |
Collapse
|
20
|
Pothast CR, Dijkland RC, Thaler M, Hagedoorn RS, Kester MGD, Wouters AK, Hiemstra PS, van Hemert MJ, Gras S, Falkenburg JHF, Heemskerk MHM. SARS-CoV-2-specific CD4 + and CD8 + T cell responses can originate from cross-reactive CMV-specific T cells. eLife 2022; 11:82050. [PMID: 36408799 PMCID: PMC9822249 DOI: 10.7554/elife.82050] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/13/2022] [Indexed: 11/22/2022] Open
Abstract
Detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) specific CD4+ and CD8+ T cells in SARS-CoV-2-unexposed donors has been explained by the presence of T cells primed by other coronaviruses. However, based on the relatively high frequency and prevalence of cross-reactive T cells, we hypothesized cytomegalovirus (CMV) may induce these cross-reactive T cells. Stimulation of pre-pandemic cryo-preserved peripheral blood mononuclear cells (PBMCs) with SARS-CoV-2 peptides revealed that frequencies of SARS-CoV-2-specific T cells were higher in CMV-seropositive donors. Characterization of these T cells demonstrated that membrane-specific CD4+ and spike-specific CD8+ T cells originate from cross-reactive CMV-specific T cells. Spike-specific CD8+ T cells recognize SARS-CoV-2 spike peptide FVSNGTHWF (FVS) and dissimilar CMV pp65 peptide IPSINVHHY (IPS) presented by HLA-B*35:01. These dual IPS/FVS-reactive CD8+ T cells were found in multiple donors as well as severe COVID-19 patients and shared a common T cell receptor (TCR), illustrating that IPS/FVS-cross-reactivity is caused by a public TCR. In conclusion, CMV-specific T cells cross-react with SARS-CoV-2, despite low sequence homology between the two viruses, and may contribute to the pre-existing immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Cilia R Pothast
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Romy C Dijkland
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Melissa Thaler
- Department of Medical Microbiology, Leiden University Medical CenterLeidenNetherlands
| | - Renate S Hagedoorn
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Michel GD Kester
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Anne K Wouters
- Department of Hematology, Leiden University Medical CenterLeidenNetherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical CenterLeidenNetherlands
| | - Martijn J van Hemert
- Department of Medical Microbiology, Leiden University Medical CenterLeidenNetherlands
| | - Stephanie Gras
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe UniversityVictoriaAustralia,Department of Biochemistry and Molecular Biology, Monash UniversityClaytonAustralia
| | | | | |
Collapse
|
21
|
Nguyen THO, Cohen CA, Rowntree LC, Bull MB, Hachim A, Kedzierska K, Valkenburg SA. T Cells Targeting SARS-CoV-2: By Infection, Vaccination, and Against Future Variants. Front Med (Lausanne) 2021; 8:793102. [PMID: 35004764 PMCID: PMC8739267 DOI: 10.3389/fmed.2021.793102] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/29/2021] [Indexed: 01/09/2023] Open
Abstract
T cell responses are a key cornerstone to viral immunity to drive high-quality antibody responses, establishing memory for recall and for viral clearance. Inefficient recruitment of T cell responses plays a role in the development of severe COVID-19 and is also represented by reduced cellular responses in men, children, and diversity compared with other epitope-specific subsets and available T cell receptor diversity. SARS-CoV-2-specific T cell responses are elicited by multiple vaccine formats and augmented by prior infection for hybrid immunity. Epitope conservation is relatively well-maintained leading to T cell crossreactivity for variants of concern that have diminished serological responses.
Collapse
Affiliation(s)
- Thi H. O. Nguyen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Carolyn A. Cohen
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Louise C. Rowntree
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Maireid B. Bull
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Asmaa Hachim
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Sophie A. Valkenburg
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- HKU-Pasteur Research Pole, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
22
|
Aleman A, Upadhyaya B, Tuballes K, Kappes K, Gleason CR, Beach K, Agte S, Srivastava K, Van Oekelen O, Barcessat V, Bhardwaj N, Kim-Schulze S, Gnjatic S, Brown B, Cordon-Cardo C, Krammer F, Merad M, Jagannath S, Wajnberg A, Simon V, Parekh S. Variable cellular responses to SARS-CoV-2 in fully vaccinated patients with multiple myeloma. Cancer Cell 2021; 39:1442-1444. [PMID: 34706273 PMCID: PMC8523488 DOI: 10.1016/j.ccell.2021.09.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Adolfo Aleman
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bhaskar Upadhyaya
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kevin Tuballes
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katerina Kappes
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charles R Gleason
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine Beach
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarita Agte
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oliver Van Oekelen
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vanessa Barcessat
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Brown
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sundar Jagannath
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ania Wajnberg
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Infectious Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Samir Parekh
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|