key: cord-0824748-2jpyjc1p authors: Casado, José L; Vizcarra, Pilar; Haemmerle, Johannes; Velasco, Héctor; Martín-Hondarza, Adrián; Rodríguez-Domínguez, Mario J.; Velasco, Tamara; Martín, Sara; Romero-Hernández, Beatriz; Fernández-Escribano, Marina; Vallejo, Alejandro title: Pre-existing T cell immunity determines the frequency and magnitude of cellular immune response to two doses of mRNA vaccine against SARS-CoV-2 date: 2022-05-02 journal: Vaccine X DOI: 10.1016/j.jvacx.2022.100165 sha: edc25d15ccadc484f1adcefeb9cdf0d5e11888cc doc_id: 824748 cord_uid: 2jpyjc1p Little is known about the factors associated with lack of T-cell response to mRNA vaccines against SARS-CoV-2. In a prospective cohort of 61 health care workers (HCWs), 21% and 16% after the first dose of mRNA BNT162b vaccine, and 12% and 7% after the second dose, showed lack of CD4(+) and CD8(+) T-cell response, respectively. Pre-existing T-cell immunity, due to past infection (46%) or cross-reactive cellular response (26%), was significantly associated with T-cell response in frequency (CD4+ T-cell, 100% vs 82% after two doses; p=0.049) and in the magnitude of T-cell response during follow up. Furthermore, baseline CD4(+) T-cell correlated positively with the titer of specific IgG-antibodies after first and second vaccine dose. Our data demonstrate that cross-reactive T-cells correlate with a better cellular response as well as an enhanced humoral response, and we confirm the close correlation of humoral and cellular response after mRNA vaccination. Current mRNA BNT162b2 vaccine reported high efficacy in preventing symptomatic SARS-CoV-2 infections after two doses [1, 2] , but the details of T-cell response following vaccination are still incompletely understood, and questions remain about the correlation between humoral and cellular response [3] . and also support the development of antibodies by B cells [4] . Moreover, T-cell responses are not significantly disrupted by the variants of concern [5] and they can contribute reducing COVID-19 severity [6] . Thus, measurement and quantification of T-cell responses will be key to identify factors associated with lack of response, to establish correlates of protection, and to understand the need of additional vaccine doses. Furthermore, pre-existing cellular response by past infection or cross-reactivity with other coronaviruses might be of importance to achieve a greater and durable immune response after vaccination [7] , an important issue since pre-existing T-cell response to SARS-CoV-2 has been observed in 30-60% of unexposed individuals [8, 9] . To clarify the differences in cellular response to the two doses of vaccine, and to identify the factors associated with a lower response both in rate and magnitude, we analyze sequentially the T-cell immune response in previously infected and uninfected health care workers (HCWs) after two doses of the Pfizer/BNT162b2 mRNA vaccine. Sixty-one HCWs evaluated 3 months before vaccination (median 147 days, IQR, 133-160) in a cross-sectional study about humoral and T-cell response to SARS-CoV-2 underwent blood analysis at least 17 days after the first and after the second dose of BNT162b2 vaccine. The participants were divided in convalescents (26, 43%) with clinical or/and serological evidence of previous SARS-CoV-2 infection, and infectionnaïve HCWs (35, 57%), who had confirmed negative serology at inclusion, and did not refer previous suggestive symptoms (fever, cough, anosmia, ageusia, headache, diarrhea) or a positive RT-PCR/serology. Both at inclusion and before vaccination, participants were tested for anti-SARS-CoV-2 IgG antibodies to SARS-CoV-2 N protein (COVID-19-SARS-CoV-2 IgG ELISA, Demeditech, Germany) to confirm serologic status and rule out subclinical infections, as this was the test used for diagnosis. After each dose of vaccination, humoral response to the S domain of the spike protein was quantified through SARS-CoV-2 IgG II Quant Alinity (Abbott, Maidenhead, UK; positivity threshold 50 arbitrary units (AU)/ml; upper limit 40,000 AU/ml). Briefly, overlapping peptides spanning the immunogenic domains of the SARS-CoV-2 spike (S) protein were used to stimulate peripheral blood mononuclear cells (PBMCs) from the participants (PepTivator SARS-CoV-2 Prot S, Miltenyi, Germany) followed by the quantitation of specific interferon (IFN)-γ-producing CD4 + and CD8 obtained from all participants. Cellular response was analyzed globally and according to the presence of prior T-cell immunity. Comparisons between groups were performed using two-tailed statistical tests, chi-square or Fisher's exact tests for categorical variables, and Mann-Whitney test or 1way analysis of variance (Kruskal-Wallis test) with Dunn's correction for multiple comparisons, as appropriate. Correlation between quantitative variables was studied using Spearman rank-order correlation test. Statistical significance was defined as twosided p values <0.05. Statistics were performed with SPSS, v 23.0. Table 1 Of note, CD4 + or CD8 + T-cell response after the first dose of vaccine were highly correlated (rho= 0.881; p<0.01). At this moment, lack of CD4 + or CD8 + T-cell response was observed in 13 (21%) and 10 (16%) individuals, respectively. Pre-existing immunity was the main factor associated with T-cell response and only 1 case of no response was observed among the 21 individuals with pre-existing immunity secondary to past infection or cross-reactivity (1/21, 5% vs 12/40, 30%; p=0.082). Thus, one third of infection-naïve HCWs without cross-reactivity had a lack of CD4 + T-cell response to the first dose of vaccine. Also, a similar lack of CD8+ T-cell response was observed in those with pre-existing immunity vs infection-naïve individuals, albeit it was not significant (1/21, 5% vs 9/40, 23%; p=0.123) Only 7 (12%) and 2 (3%) individuals remained with lack of S-specific CD4 + or CD8 + Tcell response after the second dose of the vaccine, respectively. However, as expected, these individuals were infection-naïve without cross-reactivity (18% and 5%, respectively) whereas all the patients with pre-existing immunity had CD4 + and CD8+ Tcell response to the second dose of the vaccine (p=0.042). Furthermore, pre-existing CD8 + and CD4 + T cell responses to S protein were further augmented by vaccination and it determines the magnitude of T-cell response after the second dose (CD8 + , p=0.011; CD4 + , p=0.005; Figure 2) . Notably, those without T-cell response to the first dose of vaccine had a lower cellular response to the second dose, which reach statistical significance for CD8 + T cells (Figure 3A) . We evaluated other clinical factors that could contribute to lack of T-cell response among those without pre-existing immunity. Thus, individuals without CD8 + T-cell response after the first dose were predominantly older (53 vs 47 years; p=0.11) and male (p=0.065) albeit it was not statistically significant. Before vaccination, pre-existing CD8 + and CD4 + were weakly but significantly correlated with specific antibodies pre-vaccination (CD8 + , rho=0.278, p=0.03) but this correlation with the humoral response was stronger after the first dose (CD4 + , rho=0.486, p<0.001). Furthermore, humoral response after the second dose continue to correlate with CD4 + Tcells at baseline (rho=0.413; p=0.004). As expected with this significant correlation, those individuals without pre-existing immunity and who did not develop T-cell response after the first dose of the vaccine had a lower titer of antibodies after the first dose (5,314 vs 14,159 AU/mL; p=0.021) and after the second dose (10,054 vs 20,322 AU/ml; p=0.024; Figure 3B ). Thus, in summary, infection-naïve individuals without pre-existing cross-reactive immunity had a significantly lower humoral response to first and second dose (p<0.01), and they had a lower rate of CD4+ and CD8+ T-cell response to two vaccine doses both in magnitude and in rate of response (30% and 23% after the first dose, 18% and 5% after second dose, respectively), in comparison with the stronger response observed in all the individuals with pre-existing immunity. We evaluated the CD4 + and CD8 + T-cell responses induced by SARS-CoV-2 mRNA vaccination in a well-studied cohort of SARS-CoV-2 naïve and recovered HCWs. Our data demonstrated the importance of pre-existing immunity, secondary to past infection or due to cross-reactivity, to determine the frequency and magnitude of T-cell response after one or two doses of vaccine [10] . In our study, 30% and 18% of individuals without pre-existing immunity showed lack of CD4 + T cell and lower magnitude of response to the first and second dose of the vaccine, a fact that could contribute to a weaker or shorter immune response [11] . This could be important, as antigen-specific memory CD4 + and CD8 + T cells are likely to be less impacted by antibody escape mutations in variant viral strains [5, 12] . A similar overall rate of no cellular response has been observed in other studies. Sahin described a 94% of T-cell response after two doses of mRNA vaccine [13] . Furthermore, in a similar study, Painter observed 67% and 85% of CD8+ T-cell response after the first and second dose of vaccine in infection-naïve individuals without T-cell response at baseline. [14] Moreover, we demonstrated that pre-existing cross-reactive T-cells correlated positively with the induction of S-IgG antibody titers after the first and second dose of the vaccine. These data are not surprising, since antigen-specific CD4 + T-cell response plays an important role in antigen-specific B cell development, maturation and survival [14, 15] , highlighting the convergent development of the humoral and cellular adaptive immunity [16] . Thus, cross-immunity may be responsible for the unexpectedly high efficacy of current vaccines even after a single dose [7] , as it has been demonstrated a role in decreasing the severity of infection [6, 17] . Also, pre-existing T-cell immunity could clarify some controversial data about the correlation between humoral and cellular response, especially in cross-sectional studies, due to the wide heterogeneity in the magnitude of individual spike-specific T cell responses [3] . Furthermore, HCWs without CD4 + T-cell response after the first dose and subsequent blunted cellular and humoral response after the second dose could be the best candidates for additional vaccine doses. We tried to identify other factors associated with lack of T-cell response. Vaccineinduced B cell and antibody responses have been noted to decrease with age [18] . Also, magnitude and quality of SARS-CoV-2 cross-reactivity declined with age, suggesting a possible role for age in decreasing response to the vaccine [10] . However, substantial ageassociated changes in the induction of antigen-specific T cell responses have not been previously observed [14] . We observed a trend for a worse response associated with older age and sex male, but we consider that we do not have enough sample size to be able to correctly assess this question. Limitations of our study include the small sample size. Also, time from initial T-cell evaluation (inclusion) to vaccination was around 3 months, and we cannot preclude changes in T-cell cross-reactive immunity during this period. In addition, asymptomatic infections and misclassification of cross-reactivity in the infection-naïve group was possible but unlikely because of the high sensitivity of repeated serological test at least in two determinations, at inclusion and pre-vaccination, and the similar humoral response observed in infection-naïve individuals with and without cross-reactivity. Finally, we did not have important data about incident infections in this cohort, and therefore we cannot establish the risk of disease associated with no response or with a weaker T-cell response. In conclusion, we demonstrate that pre-existing T-cells correlate with a better cellular response as well as an enhanced humoral response. Both T-cell response and humoral response were correlated following mRNA vaccination, and those infection-naïve HCWs without cellular immune response to the first dose had a weak cellular and humoral response after two doses. It remains to be determined the specific T cell response that can protect individuals against COVID-19. Also, further studies should determine the duration of clinical protection in both convalescent and infection-naïve individuals. ☒ The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. ☐The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: -Lack of CD4+ T-cell response to first and second dose of mRNA vaccine was observed in 21% and 12% of non-immunosuppressed health care workers (HCWs), respectively. -T-cell response was significantly greater in frequency and magnitude in those HCWs with pre-existing immunity, secondary to past infection or cross-reactivity. -Pre-existing immunity and T-cell response correlated with the magnitude of specific antibodies production after the first and the second dose of vaccine, and could identify the need of additional doses. Pre-existing T cell immunity determines the frequency and magnitude of cellular immune response to two doses of mRNA vaccine against SARS-CoV-2 Hospital Universitario Ramon y Cajal Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine RNA-Based COVID-19 Vaccine BNT162b2 Selected for a Pivotal Efficacy Study. medRxiv 2020 Discordant neutralizing antibody and T cell responses in asymptomatic and mild SARS-CoV-2 infection T cells in COVID-19 -united in diversity Impact of SARS-CoV-2 variants on the total CD4(+) and CD8(+) T cell reactivity in infected or vaccinated individuals SARS CoV-2 infections in healthcare workers with a pre-existing T-cell response: a prospective cohort study T-cell response after first dose of CoV-2 vaccine among healthcare workers with previous infection or cross-reactive immunity Targets of T Cell Responses to SARS CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls Cross-reactive CD4(+) T cells enhance SARS CoV-2 immune responses upon infection and vaccination A single dose of the SARS-CoV-2 vaccine BNT162b2 elicits Fc-mediated antibody effector functions and T cell responses SARS-CoV-2 variants of concern partially escape humoral but not T-cell responses in COVID-19 convalescent donors and vaccinees COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses Rapid induction of antigen-specific CD4(+) T cells is associated with coordinated humoral and cellular immunity to SARS-CoV-2 mRNA vaccination Broad and strong memory CD4 (+) and CD8 T cells induced by SARS-CoV-2 in UK convalescent COVID-19 patients Selective CD4+ T cell help for antibody responses to a large viral pathogen: deterministic linkage of specificities Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients mRNA Vaccination Induces Durable Immune Memory to SARS-CoV-2 with Continued Evolution to Variants of Concern Héctor Velasco 3 , Adrián Martín-Hondarza 3 *These authors contribute equally to this work and share first authorship 5. Department of Infectious Diseases Hospital Universitario Ramon Little is known about the factors associated with lack of T-cell response to mRNA vaccines against SARS-CoV-2. In a prospective cohort of 61 health care workers (HCWs), 21% and 16% after the first dose of mRNA BNT162b vaccine, and 12% and 7% after the second dose was significantly associated with T-cell response in frequency (CD4+ Tcell, 100% vs 82% after two doses; p=0.049) and in the magnitude of T-cell response during follow up. Furthermore, baseline CD4 + T-cell correlated positively with the titer of specific IgG-antibodies after first and second vaccine dose. Our data demonstrate that cross-reactive T-cells correlate with a better cellular response as well as an enhanced humoral response We would like to thank the study participants for their generosity in making the study possible and to Mrs. Ana Abad for her collaboration in database management. The study was funded, in part, by a grant from the Instituto de Salud Carlos III (ISCIII COV-20-01304), and department discretionary funds available to JLC. All authors attest they meet the ICMJE criteria for authorship. The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request. All the authors declare no competing interests.