Adaptive immunity and neutralizing antibodies against SARS-CoV-2 variants of concern following vaccination in patients with cancer: The CAPTURE study

Annika Fendler, Scott T C Shepherd, Lewis Au, Katalin A Wilkinson, Mary Wu, Fiona Byrne, Maddalena Cerrone, Andreas M Schmitt, Nalinie Joharatnam-Hogan, Benjamin Shum, Zayd Tippu, Karolina Rzeniewicz, Laura Amanda Boos, Ruth Harvey, Eleanor Carlyle, Kim Edmonds, Lyra Del Rosario, Sarah Sarker, Karla Lingard, Mary Mangwende, Lucy Holt, Hamid Ahmod, Justine Korteweg, Tara Foley, Jessica Bazin, William Gordon, Taja Barber, Andrea Emslie-Henry, Wenyi Xie, Camille L Gerard, Daqi Deng, Emma C Wall, Ana Agua-Doce, Sina Namjou, Simon Caidan, Mike Gavrielides, James I MacRae, Gavin Kelly, Kema Peat, Denise Kelly, Aida Murra, Kayleigh Kelly, Molly O'Flaherty, Lauren Dowdie, Natalie Ash, Firza Gronthoud, Robyn L Shea, Gail Gardner, Darren Murray, Fiona Kinnaird, Wanyuan Cui, Javier Pascual, Simon Rodney, Justin Mencel, Olivia Curtis, Clemency Stephenson, Anna Robinson, Bhavna Oza, Sheima Farag, Isla Leslie, Aljosja Rogiers, Sunil Iyengar, Mark Ethell, Christina Messiou, David Cunningham, Ian Chau, Naureen Starling, Nicholas Turner, Liam Welsh, Nicholas van As, Robin L Jones, Joanne Droney, Susana Banerjee, Kate C Tatham, Mary O'Brien, Kevin Harrington, Shreerang Bhide, Alicia Okines, Alison Reid, Kate Young, Andrew J S Furness, Lisa Pickering, Charles Swanton, Crick COVID19 consortium, Sonia Gandhi, Steve Gamblin, David Lv Bauer, George Kassiotis, Sacheen Kumar, Nadia Yousaf, Shaman Jhanji, Emma Nicholson, Michael Howell, Susanna Walker, Robert J Wilkinson, James Larkin, Samra Turajlic, Annika Fendler, Scott T C Shepherd, Lewis Au, Katalin A Wilkinson, Mary Wu, Fiona Byrne, Maddalena Cerrone, Andreas M Schmitt, Nalinie Joharatnam-Hogan, Benjamin Shum, Zayd Tippu, Karolina Rzeniewicz, Laura Amanda Boos, Ruth Harvey, Eleanor Carlyle, Kim Edmonds, Lyra Del Rosario, Sarah Sarker, Karla Lingard, Mary Mangwende, Lucy Holt, Hamid Ahmod, Justine Korteweg, Tara Foley, Jessica Bazin, William Gordon, Taja Barber, Andrea Emslie-Henry, Wenyi Xie, Camille L Gerard, Daqi Deng, Emma C Wall, Ana Agua-Doce, Sina Namjou, Simon Caidan, Mike Gavrielides, James I MacRae, Gavin Kelly, Kema Peat, Denise Kelly, Aida Murra, Kayleigh Kelly, Molly O'Flaherty, Lauren Dowdie, Natalie Ash, Firza Gronthoud, Robyn L Shea, Gail Gardner, Darren Murray, Fiona Kinnaird, Wanyuan Cui, Javier Pascual, Simon Rodney, Justin Mencel, Olivia Curtis, Clemency Stephenson, Anna Robinson, Bhavna Oza, Sheima Farag, Isla Leslie, Aljosja Rogiers, Sunil Iyengar, Mark Ethell, Christina Messiou, David Cunningham, Ian Chau, Naureen Starling, Nicholas Turner, Liam Welsh, Nicholas van As, Robin L Jones, Joanne Droney, Susana Banerjee, Kate C Tatham, Mary O'Brien, Kevin Harrington, Shreerang Bhide, Alicia Okines, Alison Reid, Kate Young, Andrew J S Furness, Lisa Pickering, Charles Swanton, Crick COVID19 consortium, Sonia Gandhi, Steve Gamblin, David Lv Bauer, George Kassiotis, Sacheen Kumar, Nadia Yousaf, Shaman Jhanji, Emma Nicholson, Michael Howell, Susanna Walker, Robert J Wilkinson, James Larkin, Samra Turajlic

Abstract

CAPTURE (NCT03226886) is a prospective cohort study of COVID-19 immunity in patients with cancer. Here we evaluated 585 patients following administration of two doses of BNT162b2 or AZD1222 vaccines, administered 12 weeks apart. Seroconversion rates after two doses were 85% and 59% in patients with solid and hematological malignancies, respectively. A lower proportion of patients had detectable neutralizing antibody titers (NAbT) against SARS-CoV-2 variants of concern (VOCs) vs wildtype (WT). Patients with hematological malignancies were more likely to have undetectable NAbT and had lower median NAbT vs solid cancers against both WT and VOCs. In comparison with individuals without cancer, patients with haematological, but not solid, malignancies had reduced NAb responses. Seroconversion showed poor concordance with NAbT against VOCs. Prior SARS-CoV-2 infection boosted NAb response including against VOCs, and anti-CD20 treatment was associated with undetectable NAbT. Vaccine-induced T-cell responses were detected in 80% of patients, and were comparable between vaccines or cancer types. Our results have implications for the management of cancer patients during the ongoing COVID-19 pandemic.

Keywords: Adaptive Immunity; Antibody Response; COVID-19; Cancer; Neutralising Antibodies; Prospective Study; SARS-CoV-2; T-cell Response; Vaccine.

Conflict of interest statement

Competing interests ST has received speaking fees from Roche, Astra Zeneca, Novartis and Ipsen. ST has the following patents filed: Indel mutations as a therapeutic target and predictive biomarker PCTGB2018/051892 and PCTGB2018/051893 and Clear Cell Renal Cell Carcinoma Biomarkers P113326GB. N.Y. has received conference support from Celegene. A.R. received a speaker fee from Merck Sharp & Dohme. J.L. has received research funding from Bristol-Myers Squibb, Merck, Novartis, Pfizer, Achilles Therapeutics, Roche, Nektar Therapeutics, Covance, Immunocore, Pharmacyclics, and Aveo, and served as a consultant to Achilles, AstraZeneca, Boston Biomedical, Bristol-Myers Squibb, Eisai, EUSA Pharma, GlaxoSmithKline, Ipsen, Imugene, Incyte, iOnctura, Kymab, Merck Serono, Nektar, Novartis, Pierre Fabre, Pfizer, Roche Genentech, Secarna, and Vitaccess. I.C. has served as a consultant to Eli-Lilly, Bristol Meyers Squibb, MSD, Bayer, Roche, Merck-Serono, Five Prime Therapeutics, Astra-Zeneca, OncXerna, Pierre Fabre, Boehringer Ingelheim, Incyte, Astella, GSK, Sotio, Eisai and has received research funding from Eli-Lilly & Janssen-Cilag. He has received honorarium from Eli-Lilly, Eisai, Servier. A.O. acknowledges receipt of research funding from Pfizer and Roche; speakers fees from Pfizer, Seagen, Lilly and AstraZeneca; is an advisory board member of Roche, Seagen, and AstraZeneca; has received conference support from Leo Pharmaceuticals, AstraZeneca/Diachi-Sankyo and Lilly. C.S. acknowledges grant support from Pfizer, AstraZeneca, Bristol Myers Squibb, Roche-Ventana, Boehringer-Ingelheim, Archer Dx Inc (collaboration in minimal residual disease sequencing technologies) and Ono Pharmaceutical, is an AstraZeneca Advisory Board member and Chief Investigator for the MeRmaiD1 clinical trial, has consulted for Amgen, Pfizer, Novartis, GlaxoSmithKline, MSD, Bristol Myers Squibb, Celgene, AstraZeneca, Illumina, Genentech, Roche-Ventana, GRAIL, Medicxi, Metabomed, Bicycle Therapeutics, and the Sarah Cannon Research Institute, has stock options in Apogen Biotechnologies, Epic Bioscience, GRAIL, and has stock options and is co-founder of Achilles Therapeutics. Patents: C.S. holds European patents relating to assay technology to detect tumour recurrence (PCT/GB2017/053289); to targeting neoantigens (PCT/EP2016/059401), identifying patent response to immune checkpoint blockade (PCT/EP2016/071471), determining HLA LOH (PCT/GB2018/052004), predicting survival rates of patients with cancer (PCT/GB2020/050221), identifying patients who respond to cancer treatment (PCT/GB2018/051912), a US patent relating to detecting tumour mutations (PCT/US2017/28013) and both a European and US patent related to identifying insertion/deletion mutation targets (PCT/GB2018/051892). L.P. has received research funding from Pierre Fabre, and honoria from Pfizer, Ipsen, Bristol-Myers Squibb, and EUSA Pharma. S.B. has recieved institutional research funding from Astrazeneca, Tesaro, GSK; speakers fees from Amgen, Pfizer, Astrazeneca, Tesaro, GSK, Clovis, Takeda, Immunogen, Mersana and has an advisor role for Amgen, Astrazeneca, Epsilogen, Genmab, Immunogen, Mersana, MSD, Merck Serono, Oncxerna, Pfizer, Roche. W.C. has received honoraria from Janssen and AstraZeneca. Remaining authors have no conflicts of interest to declare.

Figures

Figure 1. Seroconversion in cancer patients after…
Figure 1. Seroconversion in cancer patients after COVID-19 vaccination
a) Sampling and analysis schema within the CAPTURE study. Baseline samples were collected immediately before the first dose. Follow-up samples were collected: 2-4 weeks post-first dose (FU1), on the day and immediately before the second dose (FU2; ie, the additional post-first dose timepoint implemented due to delayed 12 week dosing interval), and 2-4 weeks post-second dose (FU3). S1-reactive antibody (i.e., seroconversion) and neutralising antibody assays were performed in all available follow-up samples from 585 patients. b) Proportion of infection naive patients (n= 328/323/256/312 patients at BL/FU1/FU2/FU3) with S1-reactive antibodies at each timepoint. Differences were analysed using Chi-Square test. p-values < 0.05 were considered significant. c) proportion of infection patients with S1-reactive Ab grouped by solid (n= 270/234/192/234 patients at BL/FU1/FU2/FU3) and haematological malignancies (n=58/89/64/78 patients at BL/FU1/FU2/FU3). Differences were analysed by the Chi-Square test. p-values < 0.05 were considered significant. Ab, antibodies; BL, baseline; FU1, 21-56 days post first-vaccine; FU2, 14-28 days prior to second-vaccine; FU3, 14-28days post second-vaccine
Figure 2. Neutralising antibodies against WT SARS-CoV-2…
Figure 2. Neutralising antibodies against WT SARS-CoV-2 and VOCs
a) NAbT in infection-naive patients were categorised as undetectable/low (<40), medium (40-256), or high (>256) are shown for WT SARS-CoV-2 and the three VOCs. Differences were analysed using Chi-Square test. p-values < 0.05 were considered significant. Numbers in the panel indicate sample numbers. b) NAbT in infection-naive patients against WT SARS-CoV-2, Alpha, Beta, and Delta VOCs. Median fold-decrease in NAbT is shown for each VOC in comparison to WT SARS-CoV-2 (n= 318/316/253/307 patients at BL/FU1/FU2/FU3). Dotted line at <40 denotes the lower limit of detection, dotted line at >2560 denotes the upper limit of detection. Violin plots denote density of data points. PointRange denotes the median and the 25 and 75 percentiles. Dots represent individual samples. Samples from individual patients are connected. Significance was tested by Kruskal Wallis test, p < 0.05 was considered significant, post-hoc test: two-sided Mann Whitney-U test with Bonferroni correction was used for pairwise comparisons. Only comparisons with the prior timepoint are denoted in the graph. c) Comparison of NAbT in infection-naive patients with solid (n= 262/232/189/232 patients at BL/FU1/FU2/FU3) vs haematological malignancies patients (n= 56/84/64/75 patients at BL/FU1/FU2/FU3). Dotted line at <40 denotes the lower limit of detection, dotted line at >2560 denotes the upper limit of detection. Violin plots denote density of data points. PointRange denotes the median and the 25 and 75 percentiles. Dots represent individual samples. Significance was tested by two-sided Wilcoxon-Mann-Whitney U test, p < 0.05 was considered significant. NAbT, neutralising antibody titre. NA, not tested. BL, baseline; FU1, 21-56 days post firstvaccine; FU2, 14-28 days prior to second-vaccine; FU3, 14-28days post second-vaccine.
Figure 3. Neutralising response against WT SARS-CoV-2…
Figure 3. Neutralising response against WT SARS-CoV-2 and VOCs by prior SARS-CoV-2 infection status and type of COVID-19 vaccine
a) Comparison of NAbT against WT SARS-CoV-2, Alpha, Beta, and Delta in patients with previous infection before vaccination vs infection naive patients post-second dose (n= 133/306 patients at BL/FU3). Significance was tested by two-sided Wilcoxon-Mann-Whitney U test, p < 0.05 was considered significant. b) Comparison of NAbT against WT SARS-CoV-2, Alpha, Beta, and Delta in infection naive (n= 318/316/253/307 patients at BL/FU1/FU2/FU3) vs patients previously infected with SARS-CoV-2 (n= 133/163/115/144 patients at BL/FU1/FU2/FU3). c) Comparison of NAbT against WT SARS-CoV-2, Alpha, Beta, and Delta in infection-naive patients receiving AZ (n= 262/246/212/229 patients at BL/FU1/FU2/FU3) vs PZ (n= 56/70/41/77 patients at BL/FU1/FU2/FU3, 1 patient with unknown vaccine type not included), and d) in patients with previous SARS-CoV-2 infection receiving AZ (n= 99/117/92/91 patients at BL/FU1/FU2/FU3) vs PZ (n=34/46/23/53) patients at BL/FU1/FU2/FU3). Dotted line at <40 denotes the lower limit of detection, dotted line at >2560 denotes the upper limit of detection. Violin plots denote density of data points. PointRange denotes the median and the 25 and 75 percentiles. Dots represent individual samples. Significance in b-d was tested by two sided Wilcoxon-Mann-Whitney U test, p < 0.05 was considered significant. AZ, AstraZeneca; NAbT, neutralising antibody titres; PZ, Pfizer; VOC, variant of concern. NA, not tested. BL, baseline; FU1, 21-56 days post first-vaccine; FU2, 14-28 days prior to second-vaccine; FU3, 1428days post second-vaccine.
Figure 4. WT SARS-CoV-2-specific T-cell responses in…
Figure 4. WT SARS-CoV-2-specific T-cell responses in cancer patients following vaccination
a) Exemplar ELISPOT illustrating WT SARS-CoV-2 specific T-cell response. PBMC were stimulated with 15-mer peptide pools spanning the S1 or S2 subunit of spike. T-cell responses represent the sum of SFU/106 PBMC after stimulation with WT S1 or S2 peptide pools. b) SFU/106 PBMC in infection-naive patients after vaccination (n= 165/195/122/160 patients at BL/FU1/FU2/FU3). Dotted line at <24 denotes the threshold for positivity. Violin plots denote density, PointRange the median and 25 and 75 percentiles. Dots represent individual samples. Samples from individual patients are connected. Significance was tested by Kruskal Wallis test, post-hoc test: two-sided Wilcoxon-Mann-Whitney U test with Bonferroni correction. Only comparisons with the prior timepoint are denoted in the graph. c) Comparison of SFU/106 PBMC in patients with (n= 70/88/49/69 patients at BL/FU1/FU2/FU3) and without prior SARS-CoV-2 infection (n= 165/195/122/160 patients at BL/FU1/FU2/FU3) and in d) patients with solid (n= 136/161/98/130 patients at BL/FU1/FU2/FU3) vs haematological malignancies (n= 29/34/24/30 patients at BL/FU1/FU2/FU3). Violin plots denote density, PointRange the median and 25 and 75 percentiles. Dots represent individual samples. Significance in c-d was tested by two-sided Wilcoxon-Mann-Whitney U test. e) Binary logistic regression of SFU per million PBMCs in patients with solid tumours vs haematological malignancies. Dots denote odds ratio (blue, positive odds ratio red, negative odds ratio); whiskers denote the IQR times 1.5. f) Comparison of SFU per million in patients with haematological malignancies and solid tumours pre-first dose and post-second dose. Dotted line at <24 denotes the lower limit of detection. Violin plots denote density. PointRange denotes the median and the 25 and 75 percentiles. Dots represent individual samples. Significance was tested by Kruskal Wallis test, p < 0.05 was considered significant, post-hoc test: two sided Wilcoxon-Mann-Whitney U test with Bonferroni correction. PBMC, peripheral blood mononuclear cells; NC, negative control; PC, positive control; SFU, spot-forming unit. BL, baseline; FU1, 21-56 days post first-vaccine; FU2, 14-28 days prior to second-vaccine; FU3, 14-28days post second-vaccine.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7612125/bin/EMS140022-f005.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7612125/bin/EMS140022-f006.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7612125/bin/EMS140022-f007.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7612125/bin/EMS140022-f008.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7612125/bin/EMS140022-f009.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7612125/bin/EMS140022-f010.jpg
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7612125/bin/EMS140022-f011.jpg

References

    1. Harvey WT, et al. SARS-CoV-2 variants, spike mutations and immune escape. Nature Reviews Microbiology. 2021;19(7):409–424.
    1. [cited 2021 20th July 2021];Confirmed cases of COVID-19 variants identified in UK. Latest updates on SARSCoV-2 variants detected in UK. 2021 16th July 2021. Available from: 2021.
    1. COVID-19: guidance on protecting people defined on medical grounds as extremely vulnerable. [cited 2021 20th July 2021];2021 Jul 20; Available from: 2021.
    1. Sheikh A, et al. SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness. Lancet. 2021;397(10293):2461–2462.
    1. Brosh-Nissimov T, et al. BNT162b2 vaccine breakthrough: clinical characteristics of 152 fully-vaccinated hospitalized COVID-19 patients in Israel. Clin Microbiol Infect. 2021
    1. Lopez Bernal J, et al. Effectiveness of the Pfizer-BioNTech and Oxford-AstraZeneca vaccines on covid-19 related symptoms, hospital admissions, and mortality in older adults in England: test negative case-control study. BMJ. 2021;373:n1088.
    1. Sung H, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians. 2021;71(3):209–249.
    1. Kuderer NM, et al. Clinical impact of COVID-19 on patients with cancer (CCC19): a cohort study. Lancet. 2020
    1. Grivas P, et al. Association of clinical factors and recent anticancer therapy with COVID-19 severity among patients with cancer: a report from the COVID-19 and Cancer Consortium. Ann Oncol. 2021;32(6):787–800.
    1. Lee LYW, et al. COVID-19 prevalence and mortality in patients with cancer and the effect of primary tumour subtype and patient demographics: a prospective cohort study. Lancet Oncol. 2020
    1. Garassino MC, et al. COVID-19 in patients with thoracic malignancies (TERAVOLT): first results of an international, registry-based, cohort study. Lancet Oncol. 2020
    1. Ribas A, et al. Priority COVID-19 vaccination for patients with cancer while vaccine supply is limited. Cancer Discovery. 2020:CD-20-1817
    1. [cited 2021 20th July 2021];Joint Committee on Vaccination and Immunisation: advice on priority groups for COVID-19 vaccination, 30 December 2020. 2021 Jan 6; Available from: 2021.
    1. Thakkar A, et al. Seroconversion rates following COVID-19 vaccination among patients with cancer. Cancer Cell. 2021
    1. Monin L, et al. Safety and immunogenicity of one versus two doses of the COVID-19 vaccine BNT162b2 for patients with cancer: interim analysis of a prospective observational study. Lancet Oncol. 2021
    1. Massarweh A, et al. Evaluation of Seropositivity Following BNT162b2 Messenger RNA Vaccination for SARS-CoV-2 in Patients Undergoing Treatment for Cancer. JAMA Oncology. 2021
    1. Addeo A, et al. Immunogenicity of SARS-CoV-2 messenger RNA vaccines in patients with cancer. Cancer Cell. 2021
    1. Herishanu Y, et al. Efficacy of the BNT162b2 mRNA COVID-19 Vaccine in Patients with Chronic Lymphocytic Leukemia. Blood. 2021
    1. Lim SH, et al. Antibody responses after SARS-CoV-2 vaccination in patients with lymphoma. Lancet Haematol. 2021;8(8):e542–e544.
    1. Roeker LE, et al. COVID-19 vaccine efficacy in patients with chronic lymphocytic leukemia. Leukemia. 2021:1–3.
    1. Maneikis K, et al. Immunogenicity of the BNT162b2 COVID-19 mRNA vaccine and early clinical outcomes in patients with haematological malignancies in Lithuania: a national prospective cohort study. Lancet Haematol. 2021;8(8):e583–e592.
    1. Ghione P, et al. Impaired humoral responses to COVID-19 vaccination in patients with lymphoma receiving B-cell directed therapies. Blood. 2021
    1. Parry H, et al. Antibody responses after first and second Covid-19 vaccination in patients with chronic lymphocytic leukaemia. Blood Cancer J. 2021;11(7):136.
    1. Ehmsen S, et al. Antibody and T cell immune responses following mRNA COVID-19 vaccination in patients with cancer. Cancer Cell. 2021
    1. Grifoni A, et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell. 2020
    1. Au L, et al. Cancer, COVID-19, and Antiviral Immunity: The CAPTURE Study. Cell. 2020;183(1):4–10.
    1. Wall EC, et al. AZD1222-induced neutralising antibody activity against SARS-CoV-2 Delta VOC. Lancet. 2021;398(10296):207–209.
    1. Wall EC, et al. Neutralising antibody activity against SARS-CoV-2 VOCs B.1.617.2 and B.1.351 by BNT162b2 vaccination. Lancet. 2021
    1. Carr EJ, et al. Neutralising antibodies after COVID-19 vaccination in UK haemodialysis patients. Lancet. 2021
    1. W.H.O.W.G.o.t.C. Characterisation and Management of C.-i. A minimal common outcome measure set for COVID-19 clinical research. Lancet Infect Dis. 2020;20(8):e192–e197.
    1. Thakkar A, et al. Patterns of seroconversion for SARS-CoV-2 IgG in patients with malignant disease and association with anticancer therapy. Nature Cancer. 2021
    1. Greenberger LM, et al. Antibody response to SARS-CoV-2 vaccines in patients with hematologic malignancies. Cancer Cell. 2021;39(8):1031–1033.
    1. Shrotri M, et al. Spike-antibody responses to ChAdOx1 and BNT162b2 vaccines by demographic and clinical factors (Virus Watch study) medRxiv. 2021:2021.05.12.21257102
    1. Earle KA, et al. Evidence for antibody as a protective correlate for COVID-19 vaccines. Vaccine. 2021;39(32):4423–4428.
    1. Wang Z, et al. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature. 2021;592(7855):616–622.
    1. Demonbreun AR, et al. Comparison of IgG and neutralizing antibody responses after one or two doses of COVID-19 mRNA vaccine in previously infected and uninfected individuals. EClinicalMedicine. 2021;38:101018.
    1. Khoury DS, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med. 2021;27(7):1205–1211.
    1. Bergwerk M, et al. Covid-19 Breakthrough Infections in Vaccinated Health Care Workers. New England Journal of Medicine. 2021
    1. Collier DA, et al. Age-related immune response heterogeneity to SARS-CoV-2 vaccine BNT162b2. Nature. 2021
    1. Lopez Bernal J, et al. Effectiveness of Covid-19 Vaccines against the B.1.617.2 (Delta) Variant. New England Journal of Medicine. 2021;385(7):585–594.
    1. Voysey M, et al. Single-dose administration and the influence of the timing of the booster dose on immunogenicity and efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine: a pooled analysis of four randomised trials. Lancet. 2021;397(10277):881–891.
    1. Parry H, et al. Extended interval BNT162b2 vaccination enhances peak antibody generation in older people. medRxiv. 2021:2021.05.15.21257017
    1. Kamar N, et al. Three Doses of an mRNA Covid-19 Vaccine in Solid-Organ Transplant Recipients. N Engl J Med. 2021
    1. Hall VG, et al. Randomized Trial of a Third Dose of mRNA-1273 Vaccine in Transplant Recipients. New England Journal of Medicine. 2021
    1. Normark J, et al. Heterologous ChAdOx1 nCoV-19 and mRNA-1273 Vaccination. N Engl J Med. 2021
    1. Barros-Martins J, et al. Immune responses against SARS-CoV-2 variants after heterologous and homologous ChAdOx1 nCoV-19/BNT162b2 vaccination. Nature Medicine. 2021
    1. Callaway E. Mix-and-match COVID vaccines trigger potent immune response. [cited 2021 20th July 2021];2021 Available from: .
    1. Spencer AJ, et al. Heterologous vaccination regimens with self-amplifying RNA and adenoviral COVID vaccines induce robust immune responses in mice. Nature Communications. 2021;12(1):2893
    1. Hill JA, et al. Immunogenicity of a heterologous COVID-19 vaccine after failed vaccination in a lymphoma patient. Cancer Cell. 2021
    1. Tarke A, et al. Impact of SARS-CoV-2 variants on the total CD4(+) and CD8(+) T cell reactivity in infected or vaccinated individuals. Cell Rep Med. 2021;2(7):100355.
    1. Bange EM, et al. CD8+ T cells contribute to survival in patients with COVID-19 and hematologic cancer. Nature Medicine. 2021
    1. Israelow B, et al. Adaptive immune determinants of viral clearance and protection in mouse models of SARS-CoV-2. bioRxiv. 2021
    1. McMahan K, et al. Correlates of protection against SARS-CoV-2 in rhesus macaques. Nature. 2021;590(7847):630–634.
    1. Apostolidis SA, et al. Altered cellular and humoral immune responses following SARS-CoV-2 mRNA vaccination in patients with multiple sclerosis on anti-CD20 therapy. medRxiv. 2021:2021.06.23.21259389
    1. Passaro A, et al. Severity of COVID-19 in patients with lung cancer: evidence and challenges. Journal for ImmunoTherapy of Cancer. 2021;9(3):e002266.
    1. de Joode K, et al. Dutch Oncology COVID-19 consortium: Outcome of COVID-19 in patients with cancer in a nationwide cohort study. European Journal of Cancer. 2020;141:171–184.
    1. Pritchard E, et al. Impact of vaccination on new SARS-CoV-2 infections in the United Kingdom. Nat Med. 2021
    1. Fessas P, et al. A molecular and preclinical comparison of the PD-1-targeted T-cell checkpoint inhibitors nivolumab and pembrolizumab. Semin Oncol. 2017;44(2):136–140.
    1. Faulkner N, et al. Reduced antibody cross-reactivity following infection with B.1.1.7 than with parental SARS-CoV-2 strains. bioRxiv. 2021:2021.03.01.433314
    1. Rihn SJ, et al. A plasmid DNA-launched SARS-CoV-2 reverse genetics system and coronavirus toolkit for COVID-19 research. PLoS Biol. 2021;19(2):e3001091.
    1. van den Brink EN, et al. Molecular and biological characterization of human monoclonal antibodies binding to the spike and nucleocapsid proteins of severe acute respiratory syndrome coronavirus. J Virol. 2005;79(3):1635–44.
    1. Adriana T, et al. Divergent trajectories of antiviral memory after SARS-Cov-2 infection. Research Square. 2021

Source: PubMed

3
Abonnere