10
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Rituximab‐treated patients with lymphoma develop strong CD8 T‐cell responses following COVID‐19 vaccination

      research-article
      1 , 2 , 3 , 2 , 3 , 4 , 4 , 2 , 3 , 5 , 6 , 2 , 3 , 6 , 4 , 3 , 4 , 4 , 7 , 8 , 9 , 10 , 11 , 11 , 3 , 12 , 4 , 13 , , 2 , 3 , , 1 ,
      British Journal of Haematology
      John Wiley and Sons Inc.
      anti‐CD20 antibody, CD8 T‐cell response, coronavirus disease 2019 (COVID‐19) vaccination, humoral response, lymphoma, severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) epitopes

      Read this article at

      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.

          Summary

          B‐cell depletion induced by anti‐cluster of differentiation 20 (CD20) monoclonal antibody (mAb) therapy of patients with lymphoma is expected to impair humoral responses to severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) vaccination, but effects on CD8 T‐cell responses are unknown. Here, we investigated humoral and CD8 T‐cell responses following two vaccinations in patients with lymphoma undergoing anti‐CD20‐mAb therapy as single agent or in combination with chemotherapy or other anti‐neoplastic agents during the last 9 months prior to inclusion, and in healthy age‐matched blood donors. Antibody measurements showed that seven of 110 patients had antibodies to the receptor‐binding domain of the SARS‐CoV‐2 Spike protein 3–6 weeks after the second dose of vaccination. Peripheral blood CD8 T‐cell responses against prevalent human leucocyte antigen (HLA) class I SARS‐CoV‐2 epitopes were determined by peptide‐HLA multimer analysis. Strong CD8 T‐cell responses were observed in samples from 20/29 patients (69%) and 12/16 (75%) controls, with similar median response magnitudes in the groups and some of the strongest responses observed in patients. We conclude that despite the absence of humoral immune responses in fully SARS‐CoV‐2‐vaccinated, anti‐CD20‐treated patients with lymphoma, their CD8 T‐cell responses reach similar frequencies and magnitudes as for controls. Patients with lymphoma on B‐cell depleting therapies are thus likely to benefit from current coronavirus disease 2019 (COVID‐19) vaccines, and development of vaccines aimed at eliciting T‐cell responses to non‐Spike epitopes might provide improved protection.

          Related collections

          Most cited references44

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

          COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T-cell responses

          An effective vaccine is needed to halt the spread of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic. Recently, we reported safety, tolerability and antibody response data from an ongoing placebo-controlled, observer-blinded phase I/II coronavirus disease 2019 (COVID-19) vaccine trial with BNT162b1, a lipid nanoparticle-formulated nucleoside-modified mRNA that encodes the receptor binding domain (RBD) of the SARS-CoV-2 spike protein1. Here we present antibody and T cell responses after vaccination with BNT162b1 from a second, non-randomized open-label phase I/II trial in healthy adults, 18-55 years of age. Two doses of 1-50 μg of BNT162b1 elicited robust CD4+ and CD8+ T cell responses and strong antibody responses, with RBD-binding IgG concentrations clearly above those seen in serum from a cohort of individuals who had recovered from COVID-19. Geometric mean titres of SARS-CoV-2 serum-neutralizing antibodies on day 43 were 0.7-fold (1-μg dose) to 3.5-fold (50-μg dose) those of the recovered individuals. Immune sera broadly neutralized pseudoviruses with diverse SARS-CoV-2 spike variants. Most participants had T helper type 1 (TH1)-skewed T cell immune responses with RBD-specific CD8+ and CD4+ T cell expansion. Interferon-γ was produced by a large fraction of RBD-specific CD8+ and CD4+ T cells. The robust RBD-specific antibody, T cell and favourable cytokine responses induced by the BNT162b1 mRNA vaccine suggest that it has the potential to protect against COVID-19 through multiple beneficial mechanisms.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity

            Limited knowledge is available on the relationship between antigen-specific immune responses and COVID-19 disease severity. We completed a combined examination of all three branches of adaptive immunity at the level of SARS-CoV-2-specific CD4+ and CD8+ T cell and neutralizing antibody responses in acute and convalescent subjects. SARS-CoV-2-specific CD4+ and CD8+ T cells were each associated with milder disease. Coordinated SARS-CoV-2-specific adaptive immune responses were associated with milder disease, suggesting roles for both CD4+ and CD8+ T cells in protective immunity in COVID-19. Notably, coordination of SARS-CoV-2 antigen-specific responses was disrupted in individuals > 65 years old. Scarcity of naive T cells was also associated with ageing and poor disease outcomes. A parsimonious explanation is that coordinated CD4+ T cell, CD8+ T cell, and antibody responses are protective, but uncoordinated responses frequently fail to control disease, with a connection between ageing and impaired adaptive immune responses to SARS-CoV-2.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer

              T cells directed against mutant neo-epitopes drive cancer immunity. However, spontaneous immune recognition of mutations is inefficient. We recently introduced the concept of individualized mutanome vaccines and implemented an RNA-based poly-neo-epitope approach to mobilize immunity against a spectrum of cancer mutations. Here we report the first-in-human application of this concept in melanoma. We set up a process comprising comprehensive identification of individual mutations, computational prediction of neo-epitopes, and design and manufacturing of a vaccine unique for each patient. All patients developed T cell responses against multiple vaccine neo-epitopes at up to high single-digit percentages. Vaccine-induced T cell infiltration and neo-epitope-specific killing of autologous tumour cells were shown in post-vaccination resected metastases from two patients. The cumulative rate of metastatic events was highly significantly reduced after the start of vaccination, resulting in a sustained progression-free survival. Two of the five patients with metastatic disease experienced vaccine-related objective responses. One of these patients had a late relapse owing to outgrowth of β2-microglobulin-deficient melanoma cells as an acquired resistance mechanism. A third patient developed a complete response to vaccination in combination with PD-1 blockade therapy. Our study demonstrates that individual mutations can be exploited, thereby opening a path to personalized immunotherapy for patients with cancer.
                Bookmark

                Author and article information

                Contributors
                fridtjol@gmail.com
                johanna.olweus@medisin.uio.no
                arnek2@gmail.com
                Journal
                Br J Haematol
                Br J Haematol
                10.1111/(ISSN)1365-2141
                BJH
                British Journal of Haematology
                John Wiley and Sons Inc. (Hoboken )
                0007-1048
                1365-2141
                21 March 2022
                21 March 2022
                : 10.1111/bjh.18149
                Affiliations
                [ 1 ] Department of Oncology Oslo University Hospital Oslo Norway
                [ 2 ] Department of Cancer Immunology, Institute for Cancer Research Oslo University Hospital Oslo Norway
                [ 3 ] Institute of Clinical Medicine University of Oslo Oslo Norway
                [ 4 ] Department of Immunology Oslo University Hospital Oslo Norway
                [ 5 ] Department of Hematology, Division of Medicine Akershus University Hospital Lørenskog Norway
                [ 6 ] Hematological Research Group, Division of Medicine Akershus University Hospital Lørenskog Norway
                [ 7 ] Department of Oncology University Hospital of North Norway Tromsø Norway
                [ 8 ] Department of Oncology St Olav University Hospital Trondheim Norway
                [ 9 ] Department of Oncology Haukeland University Hospital Bergen Norway
                [ 10 ] Department of Oncology and Hematology Stavanger University Hospital Stavanger Norway
                [ 11 ] Department of Neurology Oslo University Hospital Oslo Norway
                [ 12 ] Division of Cancer Medicine Oslo University Hospital Oslo Norway
                [ 13 ] ImmunoLingo Convergence Center University of Oslo Oslo Norway
                Author notes
                [*] [* ] Correspondence

                Fridtjof Lund‐Johansen, Department of Immunology, Oslo University Hospital, Oslo, Norway.

                Email: fridtjol@ 123456gmail.com

                Johanna Olweus, Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.

                Email: johanna.olweus@ 123456medisin.uio.no

                Arne Kolstad, Department of Oncology, Oslo University Hospital, Oslo, Norway.

                Email: arnek2@ 123456gmail.com

                Author information
                https://orcid.org/0000-0002-0024-0093
                Article
                BJH18149 BJH-2022-00039.R1
                10.1111/bjh.18149
                9111866
                35254660
                c7e63ed9-a876-42cb-9cce-b7f54b1a9053
                © 2022 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

                History
                : 25 February 2022
                : 07 January 2022
                : 05 March 2022
                Page count
                Figures: 3, Tables: 4, Pages: 12, Words: 6306
                Funding
                Funded by: Oslo University Hospital
                Funded by: South‐Eastern Regional Health Authority Norway
                Funded by: The Coalition for Epidemic Preparedness Innovations
                Funded by: The Norwegian Cancer Society
                Funded by: Research Council of Norway , doi 10.13039/501100005416;
                Categories
                Research Paper
                Research Papers
                Custom metadata
                2.0
                corrected-proof
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.1.6 mode:remove_FC converted:17.05.2022

                Hematology
                anti‐cd20 antibody,cd8 t‐cell response,coronavirus disease 2019 (covid‐19) vaccination,humoral response,lymphoma,severe acute respiratory syndrome coronavirus‐2 (sars‐cov‐2) epitopes

                Comments

                Comment on this article