72
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      T‐cell responses to SARS‐CoV‐2 Omicron spike epitopes with mutations after the third booster dose of an inactivated vaccine

      brief-report

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The rapidly spreading severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) Omicron variant contains more than 30 mutations that mediate escape from antibody responses elicited by prior infection or current vaccines. Fortunately, T‐cell responses are highly conserved in most individuals, but the impacts of mutations are not clear. Here, we showed that the T‐cell responses of individuals who underwent booster vaccination with CoronaVac were largely protective against the SARS‐CoV‐2 Omicron spike protein. To specifically estimate the impact of Omicron mutations on vaccinated participants, 16 peptides derived from the spike protein of the ancestral virus or Omicron strain with mutations were used to stimulate peripheral blood mononuclear cells (PBMCs) from the volunteers. Compared with the administration of two doses of vaccine, booster vaccination substantially enhanced T‐cell activation in response to both the ancestral and Omicron epitopes, although the enhancement was slightly weakened by the Omicron mutations. Then, the peptides derived from these spike proteins were used separately to stimulate PBMCs. Interestingly, compared with the ancestral peptides, only the peptides with the G339D or N440K mutation were detected to significantly destabilize the T‐cell response. Although more participants need to be evaluated to confirm this conclusion, our study nonetheless estimates the impacts of mutations on T‐cell responses to the SARS‐CoV‐2 Omicron variant.

          Related collections

          Most cited references33

          • Record: found
          • Abstract: found
          • Article: not found

          Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals

          Summary Understanding adaptive immunity to SARS-CoV-2 is important for vaccine development, interpreting coronavirus disease 2019 (COVID-19) pathogenesis, and calibration of pandemic control measures. Using HLA class I and II predicted peptide ‘megapools’, circulating SARS-CoV-2−specific CD8+ and CD4+ T cells were identified in ∼70% and 100% of COVID-19 convalescent patients, respectively. CD4+ T cell responses to spike, the main target of most vaccine efforts, were robust and correlated with the magnitude of the anti-SARS-CoV-2 IgG and IgA titers. The M, spike and N proteins each accounted for 11-27% of the total CD4+ response, with additional responses commonly targeting nsp3, nsp4, ORF3a and ORF8, among others. For CD8+ T cells, spike and M were recognized, with at least eight SARS-CoV-2 ORFs targeted. Importantly, we detected SARS-CoV-2−reactive CD4+ T cells in ∼40-60% of unexposed individuals, suggesting cross-reactive T cell recognition between circulating ‘common cold’ coronaviruses and SARS-CoV-2.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Robust T cell immunity in convalescent individuals with asymptomatic or mild COVID-19

            Summary SARS-CoV-2-specific memory T cells will likely prove critical for long-term immune protection against COVID-19. We here systematically mapped the functional and phenotypic landscape of SARS-CoV-2-specific T cell responses in unexposed individuals, exposed family members, and individuals with acute or convalescent COVID-19. Acute phase SARS-CoV-2-specific T cells displayed a highly activated cytotoxic phenotype that correlated with various clinical markers of disease severity, whereas convalescent phase SARS-CoV-2-specific T cells were polyfunctional and displayed a stem-like memory phenotype. Importantly, SARS-CoV-2-specific T cells were detectable in antibody-seronegative exposed family members and convalescent individuals with a history of asymptomatic and mild COVID-19. Our collective dataset shows that SARS-CoV-2 elicits robust, broad and highly functional memory T cell responses, suggesting that natural exposure or infection may prevent recurrent episodes of severe COVID-19.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Omicron escapes the majority of existing SARS-CoV-2 neutralizing antibodies

              The SARS-CoV-2 B.1.1.529 (Omicron) variant contains 15 mutations of the receptor-binding domain (RBD). How Omicron evades RBD-targeted neutralizing antibodies requires immediate investigation. Here we use high-throughput yeast display screening 1,2 to determine the profiles of RBD escaping mutations for 247 human anti-RBD neutralizing antibodies and show that the neutralizing antibodies can be classified by unsupervised clustering into six epitope groups (A–F)—a grouping that is highly concordant with knowledge-based structural classifications 3–5 . Various single mutations of Omicron can impair neutralizing antibodies of different epitope groups. Specifically, neutralizing antibodies in groups A–D, the epitopes of which overlap with the ACE2-binding motif, are largely escaped by K417N, G446S, E484A and Q493R. Antibodies in group E (for example, S309) 6 and group F (for example, CR3022) 7 , which often exhibit broad sarbecovirus neutralizing activity, are less affected by Omicron, but a subset of neutralizing antibodies are still escaped by G339D, N440K and S371L. Furthermore, Omicron pseudovirus neutralization showed that neutralizing antibodies that sustained single mutations could also be escaped, owing to multiple synergetic mutations on their epitopes. In total, over 85% of the tested neutralizing antibodies were escaped by Omicron. With regard to neutralizing-antibody-based drugs, the neutralization potency of LY-CoV016, LY-CoV555, REGN10933, REGN10987, AZD1061, AZD8895 and BRII-196 was greatly undermined by Omicron, whereas VIR-7831 and DXP-604 still functioned at a reduced efficacy. Together, our data suggest that infection with Omicron would result in considerable humoral immune evasion, and that neutralizing antibodies targeting the sarbecovirus conserved region will remain most effective. Our results inform the development of antibody-based drugs and vaccines against Omicron and future variants.
                Bookmark

                Author and article information

                Contributors
                didiaodexiaohuo@163.com
                binsu@ccmu.edu.cn
                Journal
                J Med Virol
                J Med Virol
                10.1002/(ISSN)1096-9071
                JMV
                Journal of Medical Virology
                John Wiley and Sons Inc. (Hoboken )
                0146-6615
                1096-9071
                06 May 2022
                06 May 2022
                : 10.1002/jmv.27814
                Affiliations
                [ 1 ] Biomedical Pioneering Innovation Center (BIOPIC) Peking University Beijing China
                [ 2 ] Beijing Advanced Innovation Center for Genomics (ICG) Peking University Beijing China
                [ 3 ] Peking‐Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies Peking University Beijing China
                [ 4 ] Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital Capital Medical University Beijing China
                Author notes
                [*] [* ] Correspondence

                Yongzheng Li, Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics (ICG), Peking‐Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.

                Email: didiaodexiaohuo@ 123456163.com

                Bin Su, Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.

                Email: binsu@ 123456ccmu.edu.cn

                [1]

                Yongzheng Li, Xiuwen Wang, and Junyan Jin contributed equally to this study.

                Author information
                http://orcid.org/0000-0001-5140-0440
                Article
                JMV27814
                10.1002/jmv.27814
                9088599
                35474581
                fc06925f-3e34-4138-9950-0c906527b406
                © 2022 Wiley Periodicals LLC.

                This article is being made freely available through PubMed Central as part of the COVID-19 public health emergency response. It can be used for unrestricted research re-use and analysis in any form or by any means with acknowledgement of the original source, for the duration of the public health emergency.

                History
                : 19 April 2022
                : 24 March 2022
                : 24 April 2022
                Page count
                Figures: 2, Tables: 0, Pages: 7, Words: 4564
                Funding
                Funded by: Ministry of Science and Technology of China
                Award ID: CPL‐1233
                Funded by: The National Natural Science Foundation of China (NSFC)
                Award ID: 81772165
                Award ID: 81974303
                Funded by: The High‐Level Public Health Specialized Talents Project of Beijing Municipal Health Commission
                Award ID: 2022‐2‐018
                Funded by: The China Primary Health Care Foundation‐Youan Medical Development Fund
                Award ID: BJYAYY‐2020PY‐01
                Funded by: The "Climbing the peak (Dengfeng)" Talent Training Program of Beijing Hospitals Authority
                Award ID: DFL20191701
                Funded by: The Beijing Health Technologies Promotion Program
                Award ID: BHTPP2020
                Funded by: Beijing Key Laboratory for HIV/AIDS Research
                Award ID: BZ0089
                Funded by: Postdoctoral Fellowship of Peking‐Tsinghua Center for Life Sciences to Y.L.
                Categories
                Short Communication
                Short Communications
                Custom metadata
                2.0
                corrected-proof
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.1.5 mode:remove_FC converted:10.05.2022

                Microbiology & Virology
                mutations,omicron,sars‐cov‐2,t‐cell responses,vaccine
                Microbiology & Virology
                mutations, omicron, sars‐cov‐2, t‐cell responses, vaccine

                Comments

                Comment on this article