Prior vaccination promotes early activation of memory T cells and enhances immune responses during SARS-CoV-2 breakthrough infection

[1]  M. Davenport,et al.  Predicting vaccine effectiveness against severe COVID-19 over time and against variants: a meta-analysis , 2023, Nature Communications.

[2]  B. Pulendran,et al.  Robust T cell responses to Pfizer/BioNTech vaccine compared to infection and evidence of attenuated peripheral CD8+ T cell responses due to COVID-19 , 2023, Immunity.

[3]  M. Davenport,et al.  SARS-CoV-2 breakthrough infection induces rapid memory and de novo T cell responses , 2023, Immunity.

[4]  D. Burton,et al.  Tissue-resident memory T cells trigger rapid exudation and local antibody accumulation , 2023, Mucosal immunology.

[5]  P. Cresswell,et al.  SARS-CoV-2 accessory proteins ORF7a and ORF3a use distinct mechanisms to down-regulate MHC-I surface expression , 2022, Proceedings of the National Academy of Sciences of the United States of America.

[6]  M. Nussenzweig,et al.  Antibody feedback regulates immune memory after SARS-CoV-2 mRNA vaccination , 2022, Nature.

[7]  D. Zeng,et al.  Association of Primary and Booster Vaccination and Prior Infection With SARS-CoV-2 Infection and Severe COVID-19 Outcomes. , 2022, JAMA.

[8]  A. Bertoletti,et al.  SARS-CoV-2 breakthrough infection in vaccinees induces virus-specific nasal-resident CD8+ and CD4+ T cells of broad specificity , 2022, The Journal of experimental medicine.

[9]  M. Nussenzweig,et al.  Memory B cell responses to Omicron subvariants after SARS-CoV-2 mRNA breakthrough infection in humans , 2022, bioRxiv.

[10]  G. Kochs,et al.  COVID-19 mRNA booster vaccine induces transient CD8+ T effector cell responses while conserving the memory pool for subsequent reactivation , 2022, Nature Communications.

[11]  L. Tserel,et al.  Protective antibodies and T cell responses to Omicron variant after the booster dose of BNT162b2 vaccine , 2022, Cell Reports Medicine.

[12]  F. Lund-Johansen,et al.  Immune responses in Omicron SARS-CoV-2 breakthrough infection in vaccinated adults , 2022, Nature Communications.

[13]  S. Honsawek,et al.  Breakthrough Infection by SARS-CoV-2 Delta and Omicron Variants Elicited Immune Response Comparable to mRNA Booster Vaccination , 2022, The Journal of infectious diseases.

[14]  M. Davenport,et al.  The magnitude and timing of recalled immunity after breakthrough infection is shaped by SARS-CoV-2 variants , 2022, Immunity.

[15]  M. Peluso,et al.  Infectious viral shedding of SARS-CoV-2 Delta following vaccination: A longitudinal cohort study , 2022, medRxiv.

[16]  L. Walker,et al.  Recall of preexisting cross-reactive B cell memory after Omicron BA.1 breakthrough infection , 2022, Science Immunology.

[17]  R. Milo,et al.  Omicron infection enhances Delta antibody immunity in vaccinated persons , 2022, Nature.

[18]  G. Kochs,et al.  SARS-CoV-2-specific T-cell epitope repertoire in convalescent and mRNA-vaccinated individuals , 2022, Nature Microbiology.

[19]  M. Nussenzweig,et al.  Increased memory B cell potency and breadth after a SARS-CoV-2 mRNA boost , 2022, Nature.

[20]  F. Tafesse,et al.  Omicron neutralizing antibody response following booster vaccination compared with breakthrough infection , 2022, Med.

[21]  O. Laeyendecker,et al.  SARS-CoV-2–specific immune responses in boosted vaccine recipients with breakthrough infections during the Omicron variant surge , 2022, JCI insight.

[22]  Justine C. Williams,et al.  Efficient recall of Omicron-reactive B cell memory after a third dose of SARS-CoV-2 mRNA vaccine , 2022, Cell.

[23]  Thomas P. Fabrizio,et al.  SARS-CoV-2 antigen exposure history shapes phenotypes and specificity of memory CD8 T cells , 2022, Nature Immunology.

[24]  L. Madoff,et al.  Characterization of immune responses in fully vaccinated individuals following breakthrough infection with the SARS-CoV-2 delta variant , 2022, Science Translational Medicine.

[25]  E. Wherry,et al.  Human epigenetic and transcriptional T cell differentiation atlas for identifying functional T cell-specific enhancers. , 2022, Immunity.

[26]  M. Koopmans,et al.  Divergent SARS CoV-2 Omicron-reactive T- and B cell responses in COVID-19 vaccine recipients , 2022, Science Immunology.

[27]  A. Sette,et al.  Development of a T cell-based immunodiagnostic system to effectively distinguish SARS-CoV-2 infection and COVID-19 vaccination status , 2022, Cell Host & Microbe.

[28]  E. Wherry,et al.  Germinal center responses to SARS-CoV-2 mRNA vaccines in healthy and immunocompromised individuals , 2022, Cell.

[29]  H. Leier,et al.  Vaccination before or after SARS-CoV-2 infection leads to robust humoral response and antibodies that effectively neutralize variants , 2022, Science Immunology.

[30]  N. Andrews,et al.  Effectiveness of COVID-19 booster vaccines against COVID-19-related symptoms, hospitalization and death in England , 2022, Nature Medicine.

[31]  S. Mallal,et al.  SARS-CoV-2 vaccination induces immunological T cell memory able to cross-recognize variants from Alpha to Omicron , 2022, Cell.

[32]  Fei Shao,et al.  Omicron escapes the majority of existing SARS-CoV-2 neutralizing antibodies , 2021, bioRxiv.

[33]  S. Haneuse,et al.  Duration of viral shedding and culture positivity with postvaccination SARS-CoV-2 delta variant infections , 2021, JCI insight.

[34]  C. Caltagirone,et al.  BNT162b2 vaccination induces durable SARS-CoV-2–specific T cells with a stem cell memory phenotype , 2021, Science Immunology.

[35]  Aaron M. Rosenfeld,et al.  mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern , 2021, Science.

[36]  E. Wherry,et al.  Rapid induction of antigen-specific CD4+ T cells is associated with coordinated humoral and cellular immunity to SARS-CoV-2 mRNA vaccination , 2021, Immunity.

[37]  H. Ljunggren,et al.  Activation and Kinetics of Circulating T Follicular Helper Cells, Specific Plasmablast Response, and Development of Neutralizing Antibodies following Yellow Fever Virus Vaccination , 2021, The Journal of Immunology.

[38]  R. Scheuermann,et al.  Impact of SARS-CoV-2 variants on the total CD4+ and CD8+ T cell reactivity in infected or vaccinated individuals , 2021, Cell Reports Medicine.

[39]  A. Mehta,et al.  Longitudinal analysis shows durable and broad immune memory after SARS-CoV-2 infection with persisting antibody responses and memory B and T cells , 2021, Cell Reports Medicine.

[40]  M. Diamond,et al.  SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses , 2021, Nature.

[41]  J. Liu,et al.  The ORF8 protein of SARS-CoV-2 mediates immune evasion through down-regulating MHC-Ι , 2021, Proceedings of the National Academy of Sciences.

[42]  M. Davenport,et al.  Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection , 2021, Nature Medicine.

[43]  Aaron M. Rosenfeld,et al.  Distinct antibody and memory B cell responses in SARS-CoV-2 naïve and recovered individuals following mRNA vaccination , 2021, Science Immunology.

[44]  S. Mallal,et al.  Comprehensive analysis of T cell immunodominance and immunoprevalence of SARS-CoV-2 epitopes in COVID-19 cases , 2021, Cell Reports Medicine.

[45]  J. Hurley,et al.  Structure of SARS-CoV-2 ORF8, a rapidly evolving immune evasion protein , 2020, Proceedings of the National Academy of Sciences.

[46]  P. Dormitzer,et al.  COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses , 2020, Nature.

[47]  J. Greenbaum,et al.  Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations with Age and Disease Severity , 2020, Cell.

[48]  Jeffrey S. Morris,et al.  SARS-CoV-2 seroprevalence among parturient women in Philadelphia , 2020, Science Immunology.

[49]  S. Hoffman,et al.  Antibody Feedback Limits the Expansion of B Cell Responses to Malaria Vaccination but Drives Diversification of the Humoral Response. , 2020, Cell host & microbe.

[50]  J. Greenbaum,et al.  Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals , 2020, Cell.

[51]  Thomas Becker,et al.  Structural basis for translational shutdown and immune evasion by the Nsp1 protein of SARS-CoV-2 , 2020, Science.

[52]  E. Harris,et al.  Neutralizing antibody titers against dengue virus correlate with protection from symptomatic infection in a longitudinal cohort , 2016, Proceedings of the National Academy of Sciences.

[53]  Paul G. Thomas,et al.  Recovery from severe H7N9 disease is associated with diverse response mechanisms dominated by CD8+ T cells , 2015, Nature Communications.

[54]  E. Wherry,et al.  Cooperativity Between CD8+ T Cells, Non-Neutralizing Antibodies, and Alveolar Macrophages Is Important for Heterosubtypic Influenza Virus Immunity , 2013, PLoS pathogens.

[55]  F. Marincola,et al.  A human memory T-cell subset with stem cell-like properties , 2011, Nature Medicine.

[56]  M. Vlková,et al.  Antibody forming cells and plasmablasts in peripheral blood in CVID patients after vaccination. , 2011, Vaccine.

[57]  E. Walsh,et al.  Peak frequencies of circulating human influenza-specific antibody secreting cells correlate with serum antibody response after immunization. , 2010, Vaccine.

[58]  S. Plotkin,et al.  Vaccines: correlates of vaccine-induced immunity. , 2008, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[59]  Ravi S. Misra,et al.  B Cells Promote Resistance to Heterosubtypic Strains of Influenza via Multiple Mechanisms1 , 2008, The Journal of Immunology.

[60]  E. Wherry,et al.  Heterogeneity and cell-fate decisions in effector and memory CD8+ T cell differentiation during viral infection. , 2007, Immunity.

[61]  F. Sallusto,et al.  Two subsets of memory T lymphocytes with distinct homing potentials and effector functions , 1999, Nature.

[62]  M. Rep,et al.  Phenotypic and Functional Separation of Memory and Effector Human CD8+ T Cells , 1997, The Journal of experimental medicine.

[63]  K. McCoy,et al.  The immune geography of IgA induction and function , 2008, Mucosal Immunology.