[en] SARS-CoV-2 vaccination significantly reduces morbidity and mortality, but has less impact on viral transmission rates, thus aiding viral evolution, and the longevity of vaccine-induced immunity rapidly declines. Immune responses in respiratory tract mucosal tissues are crucial for early control of infection, and can generate long-term antigen-specific protection with prompt recall responses. However, currently approved SARS-CoV-2 vaccines are not amenable to adequate respiratory mucosal delivery, particularly in the upper airways, which could account for the high vaccine breakthrough infection rates and limited duration of vaccine-mediated protection. In view of these drawbacks, we outline a strategy that has the potential to enhance both the efficacy and durability of existing SARS-CoV-2 vaccines, by inducing robust memory responses in the upper respiratory tract (URT) mucosa.
Disciplines :
Immunology & infectious disease
Author, co-author :
Fraser, Rupsha ; The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK. Electronic address: rupsha.fraser@ed.ac.uk
Orta-Resendiz, Aurelio; Institut Pasteur, Université Paris Cité, HIV, Inflammation and Persistence Unit, F-75015 Paris, France
MAZEIN, Alexander ; University of Luxembourg > Luxembourg Centre for Systems Biomedicine (LCSB) > Bioinformatics Core
Dockrell, David H; The University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
External co-authors :
yes
Language :
English
Title :
Upper respiratory tract mucosal immunity for SARS-CoV-2 vaccines.
We are grateful to the members of the COVID-19 Disease Map Project and DRAGON Project, which are large-scale international, collaborative efforts to describe SARS-CoV-2 virus–host interaction mechanisms and for future pandemics scenarios planning, respectively, and this review was prepared with a motivation to support these projects. R.F. A.O-R. and A.M. declare no competing interests. D.H.D. reports participation in Data and Safety Monitoring Boards for COV HIC001, COV HIC002, and Oxford SARS-CoV-2 CHIM study in seropositive volunteers, and acts as Commissioner for Medicines and Healthcare products Regulatory Agency (MHRA), UK.
Zhao, J., et al. Airway memory CD4+ T cells mediate protective immunity against emerging respiratory coronaviruses. Immunity 44 (2016), 1379–1391.
Loske, J., et al. Pre-activated antiviral innate immunity in the upper airways controls early SARS-CoV-2 infection in children. Nat. Biotechnol. 40 (2022), 319–324.
Cheemarla, N.R., et al. Dynamic innate immune response determines susceptibility to SARS-CoV-2 infection and early replication kinetics. J. Exp. Med., 218, 2021, e20210583.
Jozwik, A., et al. RSV-specific airway resident memory CD8+ T cells and differential disease severity after experimental human infection. Nat. Commun., 6, 2015, 10224.
Polack, F.P., et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N. Engl. J. Med. 383 (2020), 2603–2615.
Baden, L.R., et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 384 (2021), 403–416.
Voysey, M., et al. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet 397 (2021), 99–111.
Sadoff, J., et al. Safety and efficacy of single-dose Ad26.COV2.S vaccine against Covid-19. N. Engl. J. Med. 384 (2021), 2187–2201.
Heath, P.T., et al. Safety and efficacy of NVX-CoV2373 Covid-19 vaccine. N. Engl. J. Med. 385 (2021), 1172–1183.
Azzi, L., et al. Mucosal immune response in BNT162b2 COVID-19 vaccine recipients. eBioMedicine, 75, 2021, 103788.
Tang, J., et al. Respiratory mucosal immunity against SARS-CoV-2 following mRNA vaccination. Sci. Immunol., 2022, eadd4853.
van Doremalen, N., et al. ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques. Nature 586 (2020), 578–582.
Liu, J., et al. Heterologous Omicron-adapted vaccine as a secondary booster promotes neutralizing antibodies against Omicron and its sub-lineages in mice. Emerg. Microbes Infect., 12, 2023, e2143283.
Chalkias, S., et al. Neutralization of omicron subvariant BA.2.75 after bivalent vaccination. N. Engl. J. Med. 387 (2022), 2194–2196.
Chalkias, S., et al. A bivalent omicron-containing booster vaccine against Covid-19. N. Engl. J. Med. 387 (2022), 1279–1291.
European Medicines Agency, ETF concludes that bivalent original/Omicron BA.4-5 mRNA vaccines may be used for primary vaccination. 2022, EMA.
Singanayagam, A., et al. Community transmission and viral load kinetics of the SARS-CoV-2 delta (B.1.617.2) variant in vaccinated and unvaccinated individuals in the UK: a prospective, longitudinal, cohort study. Lancet Infect. Dis. 22 (2021), 183–195.
Acharya, C.B., et al. Viral load among vaccinated and unvaccinated, asymptomatic and symptomatic persons infected with the SARS-CoV-2 delta variant. Open Forum Infect. Dis., 9, 2022, ofac135.
Franco-Paredes, C., Transmissibility of SARS-CoV-2 among fully vaccinated individuals. Lancet Infect. Dis., 22, 2022, 16.
Tarag, Y., Stop calling it a pandemic of the unvaccinated. The Atlantic 21 September. 2021.
Rella, S.A., et al. Rates of SARS-CoV-2 transmission and vaccination impact the fate of vaccine-resistant strains. Sci. Rep., 11, 2021, 15729.
Read, A.F., et al. Imperfect vaccination can enhance the transmission of highly virulent pathogens. PLoS Biol., 13, 2015, e1002198.
Stamatatos, L., et al. mRNA vaccination boosts cross-variant neutralizing antibodies elicited by SARS-CoV-2 infection. Science 372 (2021), 1413–1418.
Emary, K.R.W., et al. Efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine against SARS-CoV-2 variant of concern 202012/01 (B.1.1.7): an exploratory analysis of a randomised controlled trial. Lancet 397 (2021), 1351–1362.
Karim, S.S.A., Vaccines and SARS-CoV-2 variants: the urgent need for a correlate of protection. Lancet 397 (2021), 1263–1264.
Sheikh, A., et al. SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness. Lancet 397 (2021), 2461–2462.
Wang, P., et al. Increased resistance of SARS-CoV-2 variant P.1 to antibody neutralization. Cell Host Microbe 29 (2021), 747–751.e4.
Wang, Z., et al. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature 592 (2021), 616–622.
Garcia-Beltran, W.F., et al. Multiple SARS-CoV-2 variants escape neutralization by vaccine-induced humoral immunity. Cell 184 (2021), 2372–2383.e9.
Christensen, P.A., et al. Delta variants of SARS-CoV-2 cause significantly increased vaccine breakthrough COVID-19 cases in Houston, Texas. Am. J. Pathol. 192 (2021), 320–331.
Venkatakrishnan, A.J., et al. Antigenic minimalism of SARS-CoV-2 is linked to surges in COVID-19 community transmission and vaccine breakthrough infections. medRxiv, 2021 Published online June 14, 2021. https://doi.org/10.1101/2021.05.23.21257668.
Riemersma, K.K., et al. Shedding of infectious SARS-CoV-2 despite vaccination. PLoS Pathog., 18, 2022, e1010876.
Chia, P.Y., et al. Virological and serological kinetics of SARS-CoV-2 Delta variant vaccine-breakthrough infections: a multi-center cohort study. Clin. Microbiol. Infect. 28 (2022), 612.e1–612.e7.
Noori, M., et al. Potency of BNT162b2 and mRNA-1273 vaccine-induced neutralizing antibodies against severe acute respiratory syndrome-CoV-2 variants of concern: a systematic review of in vitro studies. Rev. Med. Virol., 2021, e2277.
Planas, D., et al. Sensitivity of infectious SARS-CoV-2 B.1.1.7 and B.1.351 variants to neutralizing antibodies. Nat. Med. 27 (2021), 917–924.
Cele, S., et al. Omicron extensively but incompletely escapes Pfizer BNT162b2 neutralization. Nature 602 (2022), 654–656.
Andrews, N., et al. Covid-19 vaccine effectiveness against the Omicron (B.1.1.529) variant. N. Engl. J. Med. 386 (2022), 1532–1546.
Lu, L., et al. Neutralization of SARS-CoV-2 Omicron variant by sera from BNT162b2 or Coronavac vaccine recipients. Clin. Infect. Dis. 75 (2021), e822–e826.
Iketani, S., et al. Antibody evasion properties of SARS-CoV-2 Omicron sublineages. Nature 604 (2022), 553–556.
Shinde, V., et al. Efficacy of NVX-CoV2373 Covid-19 vaccine against the B.1.351 variant. N. Engl. J. Med. 384 (2021), 1899–1909.
Scheaffer, S.M., et al. Bivalent SARS-CoV-2 mRNA vaccines increase breadth of neutralization and protect against the BA.5 Omicron variant in mice. Nat. Med. 29 (2023), 247–257.
Addo, I.Y., et al. Duration of immunity following full vaccination against SARS-CoV-2: a systematic review. Arch. Public Health, 80, 2022, 200.
Goldberg, Y., et al. Protection and waning of natural and hybrid immunity to SARS-CoV-2. N. Engl. J. Med. 386 (2022), 2201–2212.
Shrotri, M., et al. Spike-antibody waning after second dose of BNT162b2 or ChAdOx1. Lancet 398 (2021), 385–387.
Pouwels, K.B., et al. Effect of Delta variant on viral burden and vaccine effectiveness against new SARS-CoV-2 infections in the UK. Nat. Med. 27 (2021), 2127–2135.
Rosenberg, E.S., et al. New COVID-19 cases and hospitalizations among adults, by vaccination status - New York, May 3-July 25, 2021. MMWR Morb. Mortal. Wkly Rep. 70 (2021), 1306–1311.
ZOE COVID Study, Mid-term effectiveness of BNT162b2 and ChAdOx1 vaccines in users of the ZOE COVID Symptom Study app in the UK. 2021.
Pérez-Alós, L., et al. Modeling of waning immunity after SARS-CoV-2 vaccination and influencing factors. Nat. Commun., 13, 2022, 1614.
Menni, C., et al. COVID-19 vaccine waning and effectiveness and side-effects of boosters: a prospective community study from the ZOE COVID Study. Lancet Infect. Dis. 22 (2022), 1002–1010.
Ferdinands, J.M., et al. Waning of vaccine effectiveness against moderate and severe Covid-19 among adults in the US from the VISION network: test negative, case-control study. BMJ, 379, 2022, e072141.
Russell, M.W., et al. Mucosal immunity in COVID-19: a neglected but critical aspect of SARS-CoV-2 infection. Front. Immunol., 11, 2020, 611337.
Nakayama, T., et al. Determinants of SARS-CoV-2 entry and replication in airway mucosal tissue and susceptibility in smokers. Cell Rep. Med., 2, 2021, 100421.
Wu, T., et al. Lung-resident memory CD8 T cells (TRM) are indispensable for optimal cross-protection against pulmonary virus infection. J. Leukoc. Biol. 95 (2014), 215–224.
Jeyanathan, M., et al. Immunological considerations for COVID-19 vaccine strategies. Nat. Rev. Immunol. 20 (2020), 615–632.
Davies, N.G., et al. Increased mortality in community-tested cases of SARS-CoV-2 lineage B.1.1.7. Nature 593 (2021), 270–274.
World Health Organization, Classification of Omicron (B.1.1.529): SARS-CoV-2 variant of concern. Published online November 26, 2021. https://www.who.int/news/item/26-11-2021-classification-of-omicron-(b.1.1.529)-sars-cov-2-variant-of-concern, 2021.
Arora, P., et al. SARS-CoV-2 Omicron sublineages show comparable cell entry but differential neutralization by therapeutic antibodies. Cell Host Microbe 30 (2022), 1103–1111.
Li, Q., et al. Antigenicity comparison of SARS-CoV-2 Omicron sublineages with other variants contained multiple mutations in RBD. MedComm (2020), 3, 2022, e130.
Reynolds, C.J., et al. Immune boosting by B.1.1.529 (Omicron) depends on previous SARS-CoV-2 exposure. Science, 2022 Published online July 15, 2022. https://doi.org/10.1126/science.abq1841.
Rochman, N., et al. Substantial impact of post-vaccination contacts on cumulative infections during viral epidemics. F1000Res., 10, 2021, 315.
P. Kearns, et al. (2021). Examining the immunological effects of COVID-19 vaccination in patients with conditions potentially leading to diminished immune response capacity – the OCTAVE Trial. Published online August 23, 2021. http://dx.doi.org/10.2139/ssrn.3910058.
Munro, C., Covid-19: 40% of patients with weakened immune system mount lower response to vaccines. BMJ, 374, 2021, n2098.
Rincon-Arevalo, H., et al. Impaired humoral immunity to SARS-CoV-2 BNT162b2 vaccine in kidney transplant recipients and dialysis patients. Sci. Immunol., 6, 2021, eabj1031.
Sattler, A., et al. Impaired humoral and cellular immunity after SARS-CoV-2 BNT162b2 (tozinameran) prime-boost vaccination in kidney transplant recipients. J. Clin. Invest., 131, 2021.
Falahi, S., Kenarkoohi, A., Host factors and vaccine efficacy: Implications for COVID-19 vaccines. J. Med. Virol. 94 (2022), 1330–1335.
Tut, G., et al. Profile of humoral and cellular immune responses to single doses of BNT162b2 or ChAdOx1 nCoV-19 vaccines in residents and staff within residential care homes (VIVALDI): an observational study. Lancet Healthy Longev. 2 (2021), e544–e553.
Pellini, R., et al. Initial observations on age, gender, BMI and hypertension in antibody responses to SARS-CoV-2 BNT162b2 vaccine. eClinicalMedicine, 36, 2021, 100928.
Ledford, H., How obesity could create problems for a COVID vaccine. Nature 586 (2020), 488–489.
Gilbert, P.B., et al. A Covid-19 milestone attained - a correlate of protection for vaccines. N. Engl. J. Med. 387 (2022), 2203–2206.
Havervall, S., et al. Anti-spike mucosal IgA protection against SARS-CoV-2 Omicron infection. N. Engl. J. Med. 387 (2022), 1333–1336.
Hassert, M., Harty, J.T., Tissue resident memory T cells — a new benchmark for the induction of vaccine-induced mucosal immunity. Front. Immunol., 13, 2022, 1039194.
Rakhra, K., et al. Exploiting albumin as a mucosal vaccine chaperone for robust generation of lung-resident memory T cells. Sci. Immunol., 6, 2021, eabd8003.
Perdomo, C., et al. Mucosal BCG vaccination induces protective lung-resident memory T cell populations against tuberculosis. mBio, 7, 2016, e01686-16.
Grau-Expósito, J., et al. Peripheral and lung resident memory T cell responses against SARS-CoV-2. Nat. Commun., 12, 2021, 3010.
Wisnewski, A.V., et al. Human IgG and IgA responses to COVID-19 mRNA vaccines. PLoS One, 16, 2021, e0249499.
Pizzolla, A., et al. Resident memory CD8+ T cells in the upper respiratory tract prevent pulmonary influenza virus infection. Sci. Immunol., 2, 2017, eaam6970.
Ishii, H., et al. Neutralizing-antibody-independent SARS-CoV-2 control correlated with intranasal-vaccine-induced CD8+ T cell responses. Cell Rep. Med., 3, 2022, 100520.
Szabo, P.A., et al. Longitudinal profiling of respiratory and systemic immune responses reveals myeloid cell-driven lung inflammation in severe COVID-19. Immunity 54 (2021), 797–814.e6.
Liao, M., et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 26 (2020), 842–844.
Slütter, B., et al. Lung airway-surveilling CXCR3hi memory CD8+ T cells are critical for protection against influenza A virus. Immunity 39 (2013), 939–948.
Netea, M.G., et al. Trained immunity: a tool for reducing susceptibility to and the severity of SARS-CoV-2 infection. Cell 181 (2020), 969–977.
Yao, Y., et al. Induction of autonomous memory alveolar macrophages requires T cell help and is critical to trained immunity. Cell 175 (2018), 1634–1650.e17.
Kar, U.K., Joosten, L.A.B., Training the trainable cells of the immune system and beyond. Nat. Immunol. 21 (2020), 115–119.
Chumakov, K., et al. Old vaccines for new infections: exploiting innate immunity to control COVID-19 and prevent future pandemics. Proc. Natl. Acad. Sci. U. S. A., 118, 2021, e2101718118.
Afkhami, S., et al. Methods and clinical development of adenovirus-vectored vaccines against mucosal pathogens. Mol. Ther. Methods Clin. Dev., 3, 2016, 16030.
Turner, D.L., et al. Lung niches for the generation and maintenance of tissue-resident memory T cells. Mucosal Immunol. 7 (2014), 501–510.
Haddadi, S., et al. Mucosal-pull induction of lung-resident memory CD8 T cells in parenteral TB vaccine-primed hosts requires cognate antigens and CD4 T cells. Front. Immunol., 10, 2019, 2075.
Bolles, M., et al. A double-inactivated severe acute respiratory syndrome coronavirus vaccine provides incomplete protection in mice and induces increased eosinophilic proinflammatory pulmonary response upon challenge. J. Virol. 85 (2011), 12201–12215.
Tseng, C.T., et al. Immunization with SARS coronavirus vaccines leads to pulmonary immunopathology on challenge with the SARS virus. PLoS One, 7, 2012, e35421.
Jeyanathan, M., et al. New tuberculosis vaccine strategies: taking aim at un-natural immunity. Trends Immunol. 39 (2018), 419–433.
Hassan, A.O., et al. An intranasal vaccine durably protects against SARS-CoV-2 variants in mice. Cell Rep., 36, 2021, 109452.
Hassan, A.O., et al. A single-dose intranasal ChAd vaccine protects upper and lower respiratory tracts against SARS-CoV-2. Cell 183 (2020), 169–184.e13.
Afkhami, S., et al. Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2. Cell 185 (2022), 896–915.e19.
Wu, S., et al. A single dose of an adenovirus-vectored vaccine provides protection against SARS-CoV-2 challenge. Nat. Commun., 11, 2020, 4081.
Aljurayyan, A., et al. Activation and induction of antigen-specific T follicular helper cells play a critical role in live-attenuated influenza vaccine-induced human mucosal anti-influenza antibody response. J. Virol. 92 (2018), e00114–e00118.
Cao, H., et al. Intranasal HD-Ad vaccine protects the upper and lower respiratory tracts of hACE2 mice against SARS-CoV-2. Cell Biosci., 11, 2021, 202.
Hartwell, B.L., et al. Intranasal vaccination with lipid-conjugated immunogens promotes antigen transmucosal uptake to drive mucosal and systemic immunity. Sci. Transl. Med., 14, 2022, eabn1413.
Ku, M.W., et al. Intranasal vaccination with a lentiviral vector protects against SARS-CoV-2 in preclinical animal models. Cell Host Microbe 29 (2021), 236–249.e6.
Li, M., et al. Mucosal vaccines: strategies and challenges. Immunol. Lett. 217 (2020), 116–125.
Alu, A., et al. Intranasal COVID-19 vaccines: from bench to bed. eBioMedicine, 76, 2022, 103841.
Chavda, V.P., et al. Intranasal vaccines for SARS-CoV-2: from challenges to potential in COVID-19 management. Drug Discov. Today 26 (2021), 2619–2636.
Lixenfeld, A.S., et al. The BioNTech / Pfizer vaccine BNT162b2 induces class-switched SARS-CoV-2-specific plasma cells and potential memory B cells as well as IgG and IgA serum and IgG saliva antibodies upon the first immunization. medRxiv, 2021 Published online March 12, 2021 https://doi.org/10.1101/2021.03.10.21252001.
Ewer, K.J., et al. T cell and antibody responses induced by a single dose of ChAdOx1 nCoV-19 (AZD1222) vaccine in a phase 1/2 clinical trial. Nat. Med. 27 (2021), 270–278.
Chan, R.W.Y., et al. The mucosal and serological immune responses to the novel coronavirus (SARS-CoV-2) vaccines. Front. Immunol., 12, 2021, 744887.
Flemming, A., Cross-reactive tissue-resident CD8+ T cells may provide first line of defence against SARS-CoV-2. Nat. Rev. Immunol., 21, 2021, 693.
Gray, J.I., et al. The roles of resident, central and effector memory CD4 T-cells in protective immunity following infection or vaccination. Immunology 154 (2018), 574–581.
Ssemaganda, A., et al. Expansion of cytotoxic tissue-resident CD8+ T cells and CCR6+CD161+ CD4+ T cells in the nasal mucosa following mRNA COVID-19 vaccination. Nat. Commun., 13, 2022, 3357.
McMaster, S.R., et al. Pulmonary antigen encounter regulates the establishment of tissue-resident CD8 memory T cells in the lung airways and parenchyma. Mucosal Immunol. 11 (2018), 1071–1078.
Corbett, K.S., et al. Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in Nonhuman Primates. N. Engl. J. Med. 383 (2020), 1544–1555.
Corbett, K.S., et al. Protection against SARS-CoV-2 beta variant in mRNA-1273 vaccine-boosted nonhuman primates. Science 374 (2021), 1343–1353.
Suh, H., et al. Microneedle patches for vaccine delivery. Clin. Exp. Vaccine Res. 3 (2014), 42–49.
Su, F., et al. Induction of mucosal immunity through systemic immunization: phantom or reality?. Hum. Vaccin. Immunother. 12 (2016), 1070–1079.
Guerrieri, M., et al. Nasal and salivary mucosal humoral immune response elicited by mRNA BNT162b2 COVID-19 vaccine compared to SARS-CoV-2 natural infection. Vaccines (Basel), 9, 2021, 1499.
Mackay, L.K., et al. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc. Natl. Acad. Sci. U. S. A. 109 (2012), 7037–7042.
Bleier, B.S., et al. COVID-19 vaccines may not prevent nasal SARS-CoV-2 infection and asymptomatic transmission. Otolaryngol. Head Neck Surg. 164 (2021), 305–307.
Lim, J.M.E., et al. SARS-CoV-2 breakthrough infection in vaccinees induces virus-specific nasal-resident CD8+ and CD4+ T cells of broad specificity. J. Exp. Med., 219, 2022.
Teijaro, J.R., Farber, D.L., COVID-19 vaccines: modes of immune activation and future challenges. Nat. Rev. Immunol. 21 (2021), 195–197.
Hemmi, M., et al. The early activation of CD8+ T cells is dependent on type I IFN signaling following intramuscular vaccination of adenovirus vector. Biomed. Res. Int., 2014, 2014, 158128.
King, C., Sprent, J., Dual nature of type I interferons in SARS-CoV-2-induced inflammation. Trends Immunol. 42 (2021), 312–322.
Madera, S., et al. Type I IFN promotes NK cell expansion during viral infection by protecting NK cells against fratricide. J. Exp. Med. 213 (2016), 225–233.
Zhu, J., et al. Type I IFN signaling on both B and CD4 T cells is required for protective antibody response to adenovirus. J. Immunol. 178 (2007), 3505–3510.
Kuka, M., et al. The role of type I interferons in CD4+ T cell differentiation. Immunol. Lett. 215 (2019), 19–23.
Knuschke, T., et al. Induction of type I interferons by therapeutic nanoparticle-based vaccination is indispensable to reinforce cytotoxic CD8+ T cell responses during chronic retroviral infection. Front. Immunol., 9, 2018, 614.
Lopez, L., et al. Dysregulated interferon response underlying severe COVID-19. Viruses, 12, 2020, 1433.
Arunachalam, P.S., et al. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science 369 (2020), 1210–1220.
Hadjadj, J., et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369 (2020), 718–724.
van Eeden, C., et al. Natural killer cell dysfunction and its role in COVID-19. Int. J. Mol. Sci., 21, 2020, 6351.
Cucak, H., et al. Type I interferon signaling in dendritic cells stimulates the development of lymph-node-resident T follicular helper cells. Immunity 31 (2009), 491–501.
Coro, E.S., et al. Type I IFN receptor signals directly stimulate local B cells early following influenza virus infection. J. Immunol. 176 (2006), 4343–4351.
Swanson, C.L., et al. Type I IFN enhances follicular B cell contribution to the T cell-independent antibody response. J. Exp. Med. 207 (2010), 1485–1500.
Le Bon, A., et al. Cutting edge: enhancement of antibody responses through direct stimulation of B and T cells by type I IFN. J. Immunol. 176 (2006), 2074–2078.
Santiago-Algarra, D., et al. Epromoters function as a hub to recruit key transcription factors required for the inflammatory response. Nat. Commun., 12, 2021, 6660.
Kamada, R., et al. Interferon stimulation creates chromatin marks and establishes transcriptional memory. Proc. Natl. Acad. Sci. U. S. A. 115 (2018), E9162–E9171.
Zhang, Q., et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science, 370, 2020, eabd4570.
Trouillet-Assant, S., et al. Type I IFN immunoprofiling in COVID-19 patients. J. Allergy Clin. Immunol. 146 (2020), 206–208.e2.
Bastard, P., et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science, 370, 2020, eabd4585.
Schroeder, S., et al. Interferon antagonism by SARS-CoV-2: a functional study using reverse genetics. Lancet Microbe 2 (2021), e210–e218.
Varese, A., et al. Type I interferons and MAVS signaling are necessary for tissue resident memory CD8+ T cell responses to RSV infection. PLoS Pathog., 18, 2022, e1010272.
Gallo, O., et al. The central role of the nasal microenvironment in the transmission, modulation, and clinical progression of SARS-CoV-2 infection. Mucosal Immunol. 14 (2021), 305–316.
Alon, R., et al. Leukocyte trafficking to the lungs and beyond: lessons from influenza for COVID-19. Nat. Rev. Immunol. 21 (2021), 49–64.
Kawamata, N., et al. Expression of endothelia and lymphocyte adhesion molecules in bronchus-associated lymphoid tissue (BALT) in adult human lung. Respir. Res., 10, 2009, 97.
Lukacs, N.W., Schaller, M., Lymphocyte trafficking and chemokine receptors during pulmonary disease. Badolato, R., Sozzani, S., (eds.) Lymphocyte Trafficking in Health and Disease, 2006, Birkhäuser Basel, 115–131.
Pejoski, D., et al. Site-specific DC surface signatures influence CD4+ T cell co-stimulation and lung-homing. Front. Immunol., 10, 2019, 1650.
Lin, Y.J., et al. Protection of hamsters challenged with SARS-CoV-2 after two doses of MVC-COV1901 vaccine followed by a single intranasal booster with nanoemulsion adjuvanted S-2P vaccine. Sci. Rep., 12, 2022, 11369.
Lin, Y., et al. Non-adjuvanted interferon-armed RBD protein nasal drops protect airway infection from SARS-CoV-2. Cell Discov., 8, 2022, 43.
Tovey, M.G., et al. Adjuvant activity of type I interferons. Biol. Chem. 389 (2008), 541–545.
Tovey, M.G., Maury, C., Oromucosal interferon therapy: marked antiviral and antitumor activity. J. Interf. Cytokine Res. 19 (1999), 145–155.
Tannir, N.M., et al. Improved tolerability and quality of life with maintained efficacy using twice-daily low-dose interferon-alpha-2b: results of a randomized phase II trial of low-dose versus intermediate-dose interferon-α-2b in patients with metastatic renal cell carcinoma. Cancer 107 (2006), 2254–2261.
Sirohi, B., et al. An open, randomized, controlled, phase II, single centre, two-period cross-over study to compare the quality of life and toxicity experienced on PEG interferon with interferon-α2b in patients with multiple myeloma maintained on a steady dose of interferon-α2b. Ann. Oncol. 18 (2007), 1388–1394.
Gary, E.N., et al. Mucosal chemokine adjuvant enhances synDNA vaccine-mediated responses to SARS-CoV-2 and provides heterologous protection in vivo. Cell Rep. Med., 3, 2022, 100693.
Caminschi, I., et al. Zymosan by-passes the requirement for pulmonary antigen encounter in lung tissue-resident memory CD8+ T cell development. Mucosal Immunol. 12 (2019), 403–412.
Jiang, R., et al. Efficacy and safety of aerosol inhalation of recombinant human interferon α1b (IFNα1b) injection for noninfluenza viral pneumonia, a multicenter, randomized, double-blind, placebo-controlled trial. J. Inflamm. (Lond.), 17, 2020, 19.
Luo, J., et al. Safety and effectiveness of inhaling different dosage recombinant human interferon α1B for bronchiolitis in children: a systematic review and meta-analysis. Evid. Based Complement. Alternat. Med., 2022, 2022, 2229735.
Zhou, Q., et al. Interferon-α92b treatment for COVID-19. Front. Immunol., 11, 2020, 1061.
de Veer, M.J., et al. Functional classification of interferon-stimulated genes identified using microarrays. J. Leukoc. Biol. 69 (2001), 912–920.
Le Bon, A., et al. Cross-priming of CD8+ T cells stimulated by virus-induced type I interferon. Nat. Immunol. 4 (2003), 1009–1015.
Schiavoni, G., et al. Type I interferons as stimulators of DC-mediated cross-priming: impact on anti-tumor response. Front. Immunol., 4, 2013, 483.
Rahimi, R.A., Luster, A.D., Chemokines: critical regulators of memory T cell development, maintenance, and function. Adv. Immunol. 138 (2018), 71–98.
Skon, C.N., et al. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat. Immunol. 14 (2013), 1285–1293.
Hondowicz, B.D., et al. IL-2 is required for the generation of viral-specific CD4+ Th1 tissue-resident memory cells and B cells are essential for maintenance in the lung. Eur. J. Immunol. 48 (2018), 80–86.
Shiow, L.R., et al. CD69 acts downstream of interferon-α/β to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 440 (2006), 540–544.
Moguche, A.O., et al. ICOS and Bcl6-dependent pathways maintain a CD4 T cell population with memory-like properties during tuberculosis. J. Exp. Med. 212 (2015), 715–728.
Hombrink, P., et al. Programs for the persistence, vigilance and control of human CD8+ lung-resident memory T cells. Nat. Immunol. 17 (2016), 1467–1478.
Holmkvist, P., et al. A major population of mucosal memory CD4+ T cells, coexpressing IL-18Rα and DR3, display innate lymphocyte functionality. Mucosal Immunol. 8 (2015), 545–558.
Hijano, D.R., et al. Type I interferon potentiates IgA immunity to respiratory syncytial virus infection during infancy. Sci. Rep., 8, 2018, 11034.
Wang, N., et al. Retrospective multicenter cohort study shows early interferon therapy is associated with favorable clinical responses in COVID-19 patients. Cell Host Microbe 28 (2020), 455–464.e2.
Park, A., Iwasaki, A., Type I and type III interferons - induction, signaling, evasion, and application to combat COVID-19. Cell Host Microbe 27 (2020), 870–878.
Bessière, P., et al. Intranasal type I interferon treatment is beneficial only when administered before clinical signs onset in the SARS-CoV-2 hamster model. PLoS Pathog., 17, 2021, e1009427.
Monk, P.D., et al. Safety and efficacy of inhaled nebulised interferon beta-1a (SNG001) for treatment of SARS-CoV-2 infection: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Respir. Med. 9 (2021), 196–206.
Channappanavar, R., et al. Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe 19 (2016), 181–193.
Dunphy, G., et al. Non-canonical activation of the DNA sensing adaptor STING by ATM and IFI16 mediates NF-κB signaling after nuclear DNA damage. Mol. Cell 71 (2018), 745–760.e5.
Berthelot, J.M., Lioté, F., COVID-19 as a STING disorder with delayed over-secretion of interferon-beta. eBioMedicine, 56, 2020, 102801.
Neufeldt, C.J., et al. SARS-CoV-2 infection induces a pro-inflammatory cytokine response through cGAS-STING and NF-κB. Commun. Biol., 5, 2020, 45.
Lee, J.S., Shin, E.C., The type I interferon response in COVID-19: implications for treatment. Nat. Rev. Immunol. 20 (2020), 585–586.
Shin, H., Iwasaki, A., A vaccine strategy that protects against genital herpes by establishing local memory T cells. Nature 491 (2012), 463–467.
Tregoning, J.S., et al. A “prime-pull” vaccine strategy has a modest effect on local and systemic antibody responses to HIV gp140 in mice. PLoS One, 8, 2013, e80559.