Durability of ChAdOx1 nCoV-19 vaccination in people living with HIV

Ane Ogbe, Matthew Pace, Mustapha Bittaye, Timothy Tipoe, Sandra Adele, Jasmini Alagaratnam, Parvinder K Aley, M Azim Ansari, Anna Bara, Samantha Broadhead, Anthony Brown, Helen Brown, Federica Cappuccini, Paola Cinardo, Wanwisa Dejnirattisai, Katie J Ewer, Henry Fok, Pedro M Folegatti, Jamie Fowler, Leila Godfrey, Anna L Goodman, Bethany Jackson, Daniel Jenkin, Mathew Jones, Stephanie Longet, Rebecca A Makinson, Natalie G Marchevsky, Moncy Mathew, Andrea Mazzella, Yama F Mujadidi, Lucia Parolini, Claire Petersen, Emma Plested, Katrina M Pollock, Thurkka Rajeswaran, Maheshi N Ramasamy, Sarah Rhead, Hannah Robinson, Nicola Robinson, Helen Sanders, Sonia Serrano, Tom Tipton, Anele Waters, Panagiota Zacharopoulou, Eleanor Barnes, Susanna Dunachie, Philip Goulder, Paul Klenerman, Gavin R Screaton, Alan Winston, Adrian Vs Hill, Sarah C Gilbert, Miles Carroll, Andrew J Pollard, Sarah Fidler, Julie Fox, Teresa Lambe, John Frater, Ane Ogbe, Matthew Pace, Mustapha Bittaye, Timothy Tipoe, Sandra Adele, Jasmini Alagaratnam, Parvinder K Aley, M Azim Ansari, Anna Bara, Samantha Broadhead, Anthony Brown, Helen Brown, Federica Cappuccini, Paola Cinardo, Wanwisa Dejnirattisai, Katie J Ewer, Henry Fok, Pedro M Folegatti, Jamie Fowler, Leila Godfrey, Anna L Goodman, Bethany Jackson, Daniel Jenkin, Mathew Jones, Stephanie Longet, Rebecca A Makinson, Natalie G Marchevsky, Moncy Mathew, Andrea Mazzella, Yama F Mujadidi, Lucia Parolini, Claire Petersen, Emma Plested, Katrina M Pollock, Thurkka Rajeswaran, Maheshi N Ramasamy, Sarah Rhead, Hannah Robinson, Nicola Robinson, Helen Sanders, Sonia Serrano, Tom Tipton, Anele Waters, Panagiota Zacharopoulou, Eleanor Barnes, Susanna Dunachie, Philip Goulder, Paul Klenerman, Gavin R Screaton, Alan Winston, Adrian Vs Hill, Sarah C Gilbert, Miles Carroll, Andrew J Pollard, Sarah Fidler, Julie Fox, Teresa Lambe, John Frater

Abstract

Duration of protection from SARS-CoV-2 infection in people living with HIV (PWH) following vaccination is unclear. In a substudy of the phase II/III the COV002 trial (NCT04400838), 54 HIV+ male participants on antiretroviral therapy (undetectable viral loads, CD4+ T cells > 350 cells/μL) received 2 doses of ChAdOx1 nCoV-19 (AZD1222) 4-6 weeks apart and were followed for 6 months. Responses to vaccination were determined by serology (IgG ELISA and Meso Scale Discovery [MSD]), neutralization, ACE-2 inhibition, IFN-γ ELISpot, activation-induced marker (AIM) assay and T cell proliferation. We show that, 6 months after vaccination, the majority of measurable immune responses were greater than prevaccination baseline but with evidence of a decline in both humoral and cell-mediated immunity. There was, however, no significant difference compared with a cohort of HIV-uninfected individuals vaccinated with the same regimen. Responses to the variants of concern were detectable, although they were lower than WT. Preexisting cross-reactive T cell responses to SARS-CoV-2 spike were associated with greater postvaccine immunity and correlated with prior exposure to beta coronaviruses. These data support the ongoing policy to vaccinate PWH against SARS-CoV-2, and they underpin the need for long-term monitoring of responses after vaccination.

Keywords: AIDS/HIV; Adaptive immunity; COVID-19; Cellular immune response; T cells.

Figures

Figure 1. PWH show higher baseline immune…
Figure 1. PWH show higher baseline immune activation and exhaustion.
(A) Schematic showing vaccination schedule for ChAdOx1 nCoV-19 in PWH. (BG) Frequency of (B) CD38+ HLA-DR+, (C) PD1+, and (D) TbetloEomeshi cells within CD4+ and (E) CD38+ HLA-DR+, (F) PD1+, and (G) TbetloEomeshi cells within CD8+ T cells. Comparison of 2 groups by 2-tailed Mann-Whitney U test. *P ≤ 0.05, ***P ≤ 0.001, and ****P ≤ 0.0001. n = 48–54 for HIV+ volunteers and 10 for HIV– control proliferation assay. Data are shown as median ± IQR.
Figure 2. Antibody levels against SARS-CoV-2 six…
Figure 2. Antibody levels against SARS-CoV-2 six months after ChAdOx1 nCoV-19 vaccination.
(AC) IgG levels for SARS-CoV-2 (A) spike, (B) RBD, and (C) N protein measured at day 0 (baseline) and day 182 (6 months after vaccination) using the MSD ELISA assays. (D) Comparison between antibody kinetics in HIV+ and HIV– across all available time points. (E and F) ACE-2 inhibition assay at baseline and 6 months after vaccination and live-virus focus reduction neutralization assay (FRNT) on n = 15 HIV+ donors on days 0, 28, 56 and 182. Comparison of 2 time points within the same group was done by Wilcoxon matched-pairs signed-rank test. Comparison of 2 groups was done by Mann-Whitney U test with Bonferroni-Dunn’s multiple-comparison test (Prism v9. B shows adjusted significant levels. **P ≤ 0.01 and ****P ≤ 0.000. Dotted lines in AC indicate cut-off points determined for each SARS-CoV-2 antigen (S, RBD, and N) based on prepandemic sera + 3 SD. n = 42–54 for HIV+ volunteers in MSD assay, in-house ELISA, and ACE-2 inhibition assay; 14–15 in FRNT assays; and 54 for HIV– controls. Data are shown as median ± IQR.
Figure 3. T cell responses following ChAdOx1…
Figure 3. T cell responses following ChAdOx1 nCoV-19 vaccination are durable in PWH.
(A) T cell response measured using peptides pools against SARS-CoV-2 S1 and S2 antigens by IFN-γ ELISpot across all time points. (B) Comparative analysis of IFN-γ T cell responses in HIV+ and HIV– volunteers. (C and D) Proliferative T cell responses to (C) SARS-CoV-2 S1 and (D) SARS-CoV-2 S2 in CD4+ T cells across all available time points. (E and F) Proliferative T cell responses to (E) SARS-CoV-2 S1 and (F) SARS-CoV-2 S2 in CD8+ T cells across all available time points. Comparison of 2 time points within the same group was done by Wilcoxon matched-pairs signed-rank test. Comparison of 2 groups was done by 2-tailed Mann-Whitney U test or multiple Mann-Whitney U test (B) with Bonferroni-Dunn’s multiple-comparison test (Prism v9). *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, and ****P ≤ 0.000. Dotted lines in CF indicate threshold for true positive based mean of DMSO controls + 3 SD. n = 48–54 for HIV+ volunteers and 54 for HIV– controls. Data are shown as median ± IQR.
Figure 4. SARS-CoV-2–specific T cells are not…
Figure 4. SARS-CoV-2–specific T cells are not preferentially biased for any CD4+ T cell subsets.
(AD) Ex vivo frequencies of (A) CXCR3+CCR6– (Th1), (B) CXCR3–CCR6– (Th2), (C) CXCR3–CCR6+ (Th17), and (D) CXCR5+ within CD4+ T cells in HIV+ volunteers measured at days 0, 42, and 182 using ex vivo T cell phenotyping. (E) Comparative analysis of frequencies of ex vivo CD4+ T cell frequencies in HIV+ and HIV– volunteers at day 182 (6 months after vaccination). (F and G) Measurement of frequencies of antigen-specific T cells including SARS-CoV-2 S1 and S2, HIV gag, and CMVpp65 using activation-induced marker (AIM) assay in (F) CD4+ and (G) CD8+ T cells. Using ‘or’ Boolean gating on FlowJo, antigen specific CD4+ T cells were: CD25+CD134(OX40)+, CD25+CD137+, or CD25+CD69+; for CD8+ T cells, antigen specific cells were: CD25+CD137+ or CD25+CD69+. (HK) Frequencies of (H) CXCR3+ CCR6– (Th1), (I) CXCR3–CCR6– (Th2), (J) CXCR3–CCR6+ (Th17), and (K) CXCR5+ CD4+ T cells within antigen-specific (AIM+) T cells in HIV+ volunteers. Comparison of 2 time points within the same group was done by Wilcoxon matched-pairs signed-rank test. Comparison of 2 groups was done by 2-tailed Mann-Whitney U test. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, and ****P ≤ 0.000. n = 48 – 54 for HIV+ volunteers in ex vivo phenotyping assay, 20 for HIV+ volunteers in AIM assay, and 10 for HIV– control in ex vivo phenotyping assay. Data are shown as median ± IQR.
Figure 5. Responses to VOCs are preserved…
Figure 5. Responses to VOCs are preserved at 6 months after ChAdOx1 nCoV-19 vaccination in PWH.
(A) ACE-2 binding inhibition assay for Alpha, Beta, and Gamma VOCs measured at day 0 (baseline) and at day 182 (6 months after vaccination) in HIV+ volunteers. (B) Comparison between ACE-2 binding inhibition of SARS-CoV-2 WT strain and Alpha, Beta, and Gamma VOCs in HIV+ volunteers. (CF) Comparison between proliferative T cell responses to SARS-CoV-2 WT strain and Beta, Gamma, and Delta VOCs in (C) CD4+ S1, (D) CD4+ S2, (E) CD8+ S1, and (F) CD8+ S2 in HIV+ volunteers. (GJ) Comparative analysis of (G) CD4+ S1, (H) CD4+ S2, (I) CD8+ S1, and (J) CD8+ S2 T cells responses to VOCs in HIV+ (solid circles) and HIV– (open circles). Comparison of 2 time points within the same group was done by Wilcoxon matched-pairs signed-rank test. Comparison of 2 groups was done by 2-tailed Mann-Whitney U test. Where indicated *P ≤ 0.05 and ****P ≤ 0.000. Dotted lines in CJ indicate threshold for true positive based mean of DMSO controls + 3 SD. n = 48–54 for ACE-2 inhibition assay in HIV+ volunteers, 20 for HIV+ VOC proliferative responses, and 10 for HIV– control VOC responses in proliferation assay. Data are shown as median ± IQR.
Figure 6. Preexisting cross-reactive CD4 + T…
Figure 6. Preexisting cross-reactive CD4+ T cell responses in PWH measured at baseline are associated with high-magnitude T cell responses after ChAdOx1 nCoV-19 vaccination.
(A and B) Baseline CD4+ SARS-CoV-2 responses were split into baseline responders (BR, proliferation > 1%, black circles and black lines) and baseline nonresponders (B-NR, proliferation < 1%, yellow circles and yellow lines), and CD4+ T cell responses after vaccination were analyzed at all available time points for (A) SARS-CoV-2 S1 and (B) SARS-CoV-2 S2. (C and D) T cells responses targeting (C) S1 and (D) S2 proteins in endemic CCCs are measured at baseline in BR and B-NR. Comparison of 2 time points within the same group was done by Wilcoxon matched-pairs signed-rank test. Comparison of 2 groups was done by 2-tailed Mann-Whitney U test with Bonferroni-Dunn’s multiple-comparison test (Prism v9). A and B show adjusted significant levels. CCC responses among participants were compared using Fisher’s exact test and listed in Supplemental Table 3. P values as indicated or *P ≤ 0.05, **P ≤ 0.01, and ****P ≤ 0.000. Dotted lines indicate threshold for true positive based mean of DMSO controls + 3 SD. n = 48–54 for HIV+ volunteers. Data are shown as median ± IQR.
Figure 7. Cross-reactive humoral immune responses among…
Figure 7. Cross-reactive humoral immune responses among Beta CoVs.
(AC) Antibody titres against (A) SARS-CoV, (B) MERS-CoV, and (C) HKU1 spike proteins measured at day 0 (baseline) and day 182 (6 months after vaccination) in HIV+ participants. (DF) Correlation between baseline antibody titres for SARS-CoV-2 and (D) SARS-CoV-1, (E) MERS-CoV, and (F) HKU1 spike protein at baseline. (G) Phylogenetic tree showing relationship between coronaviruses. Correlation was performed via Spearman’s rank correlation coefficient, and comparison of 2 time points within the same group was done by Wilcoxon matched-pairs signed-rank test. ****P ≤ 0.0001. Dotted lines in A and B indicate cut-off points determined for each SARS-CoV-2 antigen based on prepandemic sera + 3 SD. n = 48–54 for HIV+ volunteers. Data are shown as median ± IQR.

References

    1. WHO. WHO Coronavirus (COVID-19) Dashboard. Updated September 17, 2021. Accessed September 17, 2021.
    1. Mellor MM, et al. Risk of adverse coronavirus disease 2019 outcomes for people living with HIV. AIDS. 2021;35(4):1–F10.
    1. Sheth AN, et al. Influenza susceptibility, severity, and shedding in HIV-infected adults: a review of the literature. Clin Infect Dis. 2011;52(2):219–227. doi: 10.1093/cid/ciq110.
    1. Abadom TR, et al. Risk factors associated with hospitalization for influenza-associated severe acute respiratory illness in South Africa: A case-population study. Vaccine. 2016;34(46):5649–5655. doi: 10.1016/j.vaccine.2016.09.011.
    1. Tesoriero JM, et al. COVID-19 outcomes among persons living with or without diagnosed HIV infection in New York State. JAMA Netw Open. 2021;4(2):e2037069. doi: 10.1001/jamanetworkopen.2020.37069.
    1. Geretti AM, et al. Outcomes of COVID-19 related hospitalization among people with HIV in the ISARIC WHO Clinical Characterization Protocol (UK): a prospective observational study. Clin Infect Dis. 2021;73(7):2095–2106.
    1. Martin GE, et al. Epigenetic features of HIV-induced T-cell exhaustion persist despite early antiretroviral therapy. Front Immunol. 2021;12:1458.
    1. British HIV Association. BHIVA Guidelines on the Use of Vaccines in HIV-Positive Adults 2015. Updated November 2015. Accessed March 7 , 2022.
    1. Kroon FP, et al. Antibody response to influenza, tetanus and pneumococcal vaccines in HIV-seropositive individuals in relation to the number of CD4+ lymphocytes. AIDS. 1994;8(4):469–476. doi: 10.1097/00002030-199404000-00008.
    1. Cole ME, et al. Responses to quadrivalent influenza vaccine reveal distinct circulating CD4+CXCR5+ T cell subsets in men living with HIV. Sci Rep. 2019;9(1):15650. doi: 10.1038/s41598-019-51961-9.
    1. Zanetti AR, et al. Safety and immunogenicity of influenza vaccination in individuals infected with HIV. Vaccine. 2002;20:B29–B32. doi: 10.1016/S0264-410X(02)00511-X.
    1. Bridges CB, et al. Prevention and control of influenza. Recommendations of the Advisory Committee on Immunization Practices (ACIP) MMWR Recomm Rep. 2001;50(rr-4):1–44.
    1. Christensen-Quick A, et al. Influenza vaccination can broadly activate the HIV reservoir during antiretroviral therapy. J Acquir Immune Defic Syndr. 2018;79(3):104–e107.
    1. Yek C, et al. Standard vaccines increase HIV-1 transcription during antiretroviral therapy. AIDS. 2016;30(15):2289–2298. doi: 10.1097/QAD.0000000000001201.
    1. Ramasamy MN, et al. Safety and immunogenicity of ChAdOx1 nCoV-19 vaccine administered in a prime-boost regimen in young and old adults (COV002): a single-blind, randomised, controlled, phase 2/3 trial. Lancet. 2020;396(10267):1979–1993. doi: 10.1016/S0140-6736(20)32466-1.
    1. Frater J, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 (AZD1222) vaccine against SARS-CoV-2 in HIV infection: a single-arm substudy of a phase 2/3 clinical trial. Lancet HIV. 2021;8(8):474–485.
    1. Folegatti PM, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet. 2020;396(10249):467–478. doi: 10.1016/S0140-6736(20)31604-4.
    1. Ewer KJ, 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. 2021;27(2):270–278. doi: 10.1038/s41591-020-01194-5.
    1. Tomic A, et al. Divergent trajectories of antiviral memory after SARS-Cov-2 infection [preprint]. Posted on Research Square June 15, 2021.
    1. Widge AT, et al. Durability of responses after SARS-CoV-2 mRNA-1273 vaccination. N Engl J Med. 2020;384(1):80–82.
    1. Mateus J, et al. Low-dose mRNA-1273 COVID-19 vaccine generates durable memory enhanced by cross-reactive T cells. Science. 2021;374(6566):eabj9853. doi: 10.1126/science.abj9853.
    1. Pegu A, et al. Durability of mRNA-1273 vaccine-induced antibodies against SARS-CoV-2 variants. Science. 2021;373(6561):1372–1377. doi: 10.1126/science.abj4176.
    1. GISAID EpiFlu Database. Freunde von GISAID e.V; 2022. Accessed March 7, 2022.
    1. Lopez Bernal J, et al. , Effectiveness of Covid-19 vaccines against the B.1.617.2 (Delta) variant. N Engl J Med. 2021;385(7):585–594. doi: 10.1056/NEJMoa2108891.
    1. Public Health England. SARS-CoV-2 variants of concern and variants under investigation in England Technical briefing 15. Updated June 11, 2021. Accessed July 18, 2021. .
    1. Nasreen S, et al. Effectiveness of COVID-19 vaccines against variants of concern in Ontario, Canada [preprint]. Posted on medRxiv July 3, 2021.
    1. Bergwerk M, et al. Covid-19 breakthrough infections in vaccinated health care workers. N Engl J Med. 2021;385(16):1474–1484. doi: 10.1056/NEJMoa2109072.
    1. Mateus J, et al. Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science. 2020;370(6512):89–94. doi: 10.1126/science.abd3871.
    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;181(7):1489–1501. doi: 10.1016/j.cell.2020.05.015.
    1. Braun J, et al. SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19. Nature. 2020;587(7833):270–274. doi: 10.1038/s41586-020-2598-9.
    1. Le Bert N, et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature. 2020;584(7821):457–462. doi: 10.1038/s41586-020-2550-z.
    1. Ng KW, et al. Preexisting and de novo humoral immunity to SARS-CoV-2 in humans. Science. 2020;370(6522):1339. doi: 10.1126/science.abe1107.
    1. Ogbe A, et al. T cell assays differentiate clinical and subclinical SARS-CoV-2 infections from cross-reactive antiviral responses. Nat Commun. 2021;12(1):2055. doi: 10.1038/s41467-021-21856-3.
    1. Swadling L, et al. Pre-existing polymerase-specific T cells expand in abortive seronegative SARS-CoV-2. Nature. 2022;601(7891):110–117. doi: 10.1038/s41586-021-04186-8.
    1. Shrock E, et al. Viral epitope profiling of COVID-19 patients reveals cross-reactivity and correlates of severity. Science. 2020;370(6520):eabd4250. doi: 10.1126/science.abd4250.
    1. Loyal L, et al. Cross-reactive CD4+ T cells enhance SARS-CoV-2 immune responses upon infection and vaccination. Science. 2021;374(6564):eabh1823. doi: 10.1126/science.abh1823.
    1. Poston D, et al. Absence of SARS-CoV-2 neutralizing activity in pre-pandemic sera from individuals with recent seasonal coronavirus infection [preprint]. Posted on medRxiv October 11, 2020.
    1. Anderson EM, et al. Seasonal human coronavirus antibodies are boosted upon SARS-CoV-2 infection but not associated with protection. Cell. 2021;184(7):1858–1864. doi: 10.1016/j.cell.2021.02.010.
    1. McNaughton AL, et al. Fatal COVID-19 outcomes are associated with an antibody response targeting epitopes shared with endemic coronaviruses [preprint]. Posted on medRxiv May 7, 2021.
    1. Hileman CO, Funderburg NT. Inflammation, immune activation, and antiretroviral therapy in HIV. Curr HIV/AIDS Rep. 2017;14(3):93–100. doi: 10.1007/s11904-017-0356-x.
    1. Nakanjako D, et al. High T-cell immune activation and immune exhaustion among individuals with suboptimal CD4 recovery after 4 years of antiretroviral therapy in an African cohort. BMC Infect Dis. 2011;11(1):43. doi: 10.1186/1471-2334-11-43.
    1. Almeida CAM, et al. Immune activation in patients infected with HIV type 1 and maintaining suppression of viral replication by highly active antiretroviral therapy. AIDS Res Hum Retroviruses. 2002;18(18):1351–1355. doi: 10.1089/088922202320935429.
    1. Madhi SA, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 (AZD1222) vaccine against SARS-CoV-2 in people living with and without HIV in South Africa: an interim analysis of a randomised, double-blind, placebo-controlled, phase 1B/2A trial. Lancet HIV. 2021;8(9):568–580.
    1. Falsey AR, et al. Phase 3 safety and efficacy of AZD1222 (ChAdOx1 nCoV-19) Covid-19 vaccine. N Engl J Med. 2021;385(25):2348–2360. doi: 10.1056/NEJMoa2105290.
    1. Kunisaki KM, Janoff EN. Influenza in immunosuppressed populations: a review of infection frequency, morbidity, mortality, and vaccine responses. Lancet Infect Dis. 2009;9(8):493–504. doi: 10.1016/S1473-3099(09)70175-6.
    1. Golding B, Scott DE. Vaccine strategies: targeting helper T cell responses. Ann N Y Acad Sci. 1995;754:126–137. doi: 10.1111/j.1749-6632.1995.tb44445.x.
    1. Nielsen CM, et al. Protein/AS01B vaccination elicits stronger, more Th2-skewed antigen-specific human T follicular helper cell responses than heterologous viral vectors. Cell Rep Med. 2021;2(3):100207. doi: 10.1016/j.xcrm.2021.100207.
    1. Taylor JM, et al. Effects of a Th1- versus a Th2-biased immune response in protection against Helicobacter pylori challenge in mice. Microb Pathog. 2008;44(1):20–27. doi: 10.1016/j.micpath.2007.06.006.
    1. Bowyer G, et al. CXCR3+ T follicular helper cells induced by co-administration of RTS,S/AS01B and viral-vectored vaccines are associated with reduced immunogenicity and efficacy against malaria. Front Immunol. 2018;9:1660. doi: 10.3389/fimmu.2018.01660.
    1. Bentebibel SE, et al. ICOS(+)PD-1(+)CXCR3(+) T follicular helper cells contribute to the generation of high-avidity antibodies following influenza vaccination. Sci Rep. 2016;6(1):26494. doi: 10.1038/srep26494.
    1. Rodda LB, et al. Functional SARS-CoV-2-specific immune memory persists after mild COVID-19. Cell. 2021;184(1):169–183. doi: 10.1016/j.cell.2020.11.029.
    1. Rydyznski Moderbacher C, et al. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell. 2020;183(4):996–1012. doi: 10.1016/j.cell.2020.09.038.
    1. Juno JA, et al. Humoral and circulating follicular helper T cell responses in recovered patients with COVID-19. Nat Med. 2020;26(9):1428–1434. doi: 10.1038/s41591-020-0995-0.
    1. Imai T, et al. Selective recruitment of CCR4-bearing Th2 cells toward antigen-presenting cells by the CC chemokines thymus and activation-regulated chemokine and macrophage-derived chemokine. Int Immunol. 1999;11(1):81–88. doi: 10.1093/intimm/11.1.81.
    1. Mikhak Z, et al. Contribution of CCR4 and CCR8 to antigen-specific T(H)2 cell trafficking in allergic pulmonary inflammation. J Allergy Clin Immunol. 2009;123(1):67–73. doi: 10.1016/j.jaci.2008.09.049.
    1. Kara EE, et al. Distinct chemokine receptor axes regulate Th9 cell trafficking to allergic and autoimmune inflammatory sites. J Immunol. 2013;191(3):1110–1117. doi: 10.4049/jimmunol.1203089.
    1. Chen T, et al. Th9 cell differentiation and its dual effects in tumor development. Front Immunol. 2020;11:1026. doi: 10.3389/fimmu.2020.01026.
    1. Zhou R, et al. Acute SARS-CoV-2 infection impairs dendritic cell and T cell responses. Immunity. 2020;53(4):864–877. doi: 10.1016/j.immuni.2020.07.026.
    1. Dan JM, et al. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science. 2021;371(6529):eabf4063. doi: 10.1126/science.abf4063.
    1. Tso FY, et al. Presence of antibody-dependent cellular cytotoxicity (ADCC) against SARS-CoV-2 in COVID-19 plasma. PLoS One. 2021;16(3):e0247640. doi: 10.1371/journal.pone.0247640.
    1. Lee WS, et al. Decay of Fc-dependent antibody functions after mild to moderate COVID-19. Cell Rep Med. 2021;2(6):100296. doi: 10.1016/j.xcrm.2021.100296.
    1. Chen X, et al. The development and kinetics of functional antibody-dependent cell-mediated cytotoxicity (ADCC) to SARS-CoV-2 spike protein. Virology. 2021;559:1–9. doi: 10.1016/j.virol.2021.03.009.
    1. Wajnberg A, et al. Robust neutralizing antibodies to SARS-CoV-2 infection persist for months. Science. 2020;370(6521):1227–1230. doi: 10.1126/science.abd7728.
    1. Alrubayyi A, et al. Characterization of humoral and SARS-CoV-2 specific T cell responses in people living with HIV. Nat Commun. 2021;12(1):5839. doi: 10.1038/s41467-021-26137-7.
    1. NHS. Coronavirus (COVID-19) Vaccines. Updated September 1, 2022. Accessed September 17, 2022.
    1. Geers D, et al. SARS-CoV-2 variants of concern partially escape humoral but not T-cell responses in COVID-19 convalescent donors and vaccinees. Sci Immunol. 2021;6(59):eabj1750. doi: 10.1126/sciimmunol.abj1750.
    1. Liu Y, et al. Neutralizing activity of BNT162b2-elicited serum. N Engl J Med. 2021;384(15):1466–1468. doi: 10.1056/NEJMc2102017.
    1. Wang P. Antibody resistance of SARS-CoV-2 variants B.1.351 and B.1.1.7. Nature. 2021;593(7857):130–135. doi: 10.1038/s41586-021-03398-2.
    1. Supasa P, et al. Reduced neutralization of SARS-CoV-2 B.1.1.7 variant by convalescent and vaccine sera. Cell. 2021;184(8):2201–2211. doi: 10.1016/j.cell.2021.02.033.

Source: PubMed

3
Subscribe