key: cord-0811494-4ah8zmlz authors: Tormo, Nuria; Navalpotro, David; Martínez-Serrano, María; Moreno, Marta; Grosson, Fernando; Tur, Irene; Guna, M Remedios; Soriano, Pepa; Tornero, Ana; Gimeno, Concepción title: Commercial Interferon-gamma release assay to assess the immune response to first and second doses of mRNA vaccine in previously COVID-19 infected versus uninfected individuals. date: 2021-10-10 journal: Diagn Microbiol Infect Dis DOI: 10.1016/j.diagmicrobio.2021.115573 sha: d7be8fa82a25ab6c53451b89c2b117564fda12e2 doc_id: 811494 cord_uid: 4ah8zmlz We analysed immunological response during vaccination by using quantitative anti-spike IgG antibodies (qAbs) and Interferon-gamma (IFNγ) production by SARS-CoV-2-specific CD4+ and CD8+ T cells (QuantiFERON(Ⓡ) assay). Blood samples were collected at four time points: a day before the reception of first (T0) and second (T1) BNT162b2 doses, 14 (T2) and 28 days (T3) after second dose. Fifty individuals were included: 34 previously infected by SARS-CoV-2 (CoV2+) and 16 that were not (CoV2-). Among CoV2+, we only observed significant differences after the first dose in both qAbs and IFNγ+ T cells. CoV2- showed differences after each dose, and the response was lower than CoV2+. Older people presented a higher response in CoV2+, while in CoV2, young people responded best. Our results suggest that the second BNT162b2 vaccine dose is not a priority in people with previous COVID-19. QuantiFERON(Ⓡ) is a good option to monitor T-cell immunity to SARS-CoV-2. The primary goal of SARS-CoV-2 vaccines is to stop transmission and prevent infection and disease, mainly focusing on those who are at higher risk of severe complications, as is the case for older people. Rapid vaccination implementation is now the world´s health care priority. The first vaccines that have been approved-BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna)-rely on a new vaccine approach, the introduction of lipid nanoparticles containing messenger RNA (mRNA) molecules encoding for the spike (S) protein of the virus, which is the main target of neutralizing antibodies (NABS) against SARS-CoV-2 [1] . Studies in non-human primates have shown that infection with wildtype viruses prevents subsequent reinfection, thus indicating the existence of protective immunity, which has been associated with the production of NABS [2] , [3] . Prevention of reinfection has also been observed in human epidemiological studies [4] , indicating that sufficient NABS may be a clue for protection against COVID-19. The first vaccine trials have demonstrated good immunogenicity and similar levels of neutralizing and receptor-binding domain (RBD) antibodies in vaccinated compared to infected individuals [1] , [5] . Some pre-clinical studies in non-human primates have shown that vaccines also elicit a T cell response, which may help protect against challenge with the virus [3] , [6] . Furthermore, the first trials in humans with mRNA vaccines have concluded that they are able to induce a Th1-polarized cell response after a 2dose schedule [7] rather than a Th2 response, which has been related to complications after SARS and MERS vaccinations in animal models [8] , [9] . What is clear is that a study of the combination of the humoral and cellular response is needed to increase knowledge of the efficacy of the vaccines-including, most importantly, whether they can protect from infection and severe cases of COVID- 19 , and if so what levels of surrogate markers may indicate protection. It has also been demonstrated that a robust CD4+ and CD8+ T cell activation is reached after natural SARS-CoV-2 infection [10] , [11] , even in the absence of circulating antibodies [10] , [12] . Moreover, an adaptive immune response can be detected long after a natural infection [13] , [14] . But, limited research has been conducted regarding the immune response, especially the T cell response, to vaccination after a previous natural infection [1] , and this situation is common, given that a large number of people have already been exposed to the virus. And, if a single dose is able to boost protection sufficiently, perhaps the vaccination schedule could be modified, as has been suggested [15] , [16] . Different assays have been used to assess T cell response to various pathogens, including viruses, by using different platforms and technologies, such as intracellular cytokine staining followed by flow cytometry, peptide-major histocompatibility complex multimer technologies, or IFNγ production detected either by ELISpot or ELISA [17] . The QuantiFERONⓇ SARS-CoV-2 RUO (Qiagen) consists of the detection of IFNγ production by either CD4+ or CD4+ and CD8+ T cells by an ELISA assay, benefiting from the previous experience with the tuberculosis assay [18] , highly expanded and standardized. Our goal is to provide data so that public health policies can implement the best vaccination strategies to optimize available vaccines and achieve high vaccination coverage quickly. Therefore, we have investigated immunological responses after the first and second BNT162b2 doses by using quantitative antispike IgG antibodies and IFNγ production by CD4+ and CD8+ T cells with the new commercial IFNγ release assay (IGRA), QuantiFERONⓇ SARS-CoV-2 RUO (Qiagen), as markers of humoral and cellular immunity, respectively. We included fifty individuals from a nursing home: 27 residents, all over 60 years old, and 23 socio-sanitary staff, all under 60 years old, scheduled for BNT162b2 vaccination in January 2021. Previous COVID-19 infection in March-April 2020 was confirmed in 34 people (CoV2+), 31 of them by RNA RT-PCR and three by seroconversion. All of the cases were classified as asymptomatic or mild, with only two of them requiring hospital admission. Serum and whole blood samples of participants were drawn at four time points: the day before the administration of the first (T0) and second (T1) BNT162b2 doses; then, 14 (T2) and 28 days (T3) after the second dose. Clinical data were not recorded as this was not the objective of the study. All the procedures of the study were carried out in accordance with the ethical standards described in the Helsinki Declaration. Confirmed consent was obtained from all participants. The study was approved by our ethics committee (140/2020). SARS-CoV-2 specific antibodies were detected with the quantitative IgG (S) enzyme-linked immunosorbent assay (ELISA) (Euroimmun, Lübeck, Germany), followed by additional 1:10 and 1:100 sample dilutions, as recommended by the manufacturer. A result >10 RU/ml is considered positive. Cellular immune response was assessed with the QuantiFERONⓇ SARS-CoV-2 RUO (Qiagen) commercial assay. This test is based on in vitro CD4+ and CD8+ lymphocytes stimulation in heparinized whole blood with a combination of specific SARS-CoV-2 antigens covering the S protein, followed by measurement in plasma of IFNγ production by ELISA (QuantiFERONⓇ ELISA). Briefly, the assay has two sets with two different tubes each, which are designed to draw 1 ml of blood per tube for stimulation. The QuantiFERON SARS-CoV-2 Starter Set Blood Collection Tubes (BCT) consist of two tubes, named Ag1 and Ag2, which detect IFNγ production by CD4+ (IFNγ+CD4+) T cells and CD4+ and CD8+ (IFNγ+CD4+&CD8+) T cells together, respectively. The Ag1 tube contains CD4+ epitopes derived from the S1 subunit (Receptor Binging Domain, RBD), and the Ag2 tube contains CD4+ and CD8+ epitopes from the S1 and S2 subunits. The Control Tube Set contains negative (Nil) and positive (Mitogen) control tubes. All tubes were incubated at 37ºC for 16-24 h, then centrifuged for 15 min at 2500 g to harvest the plasma. Plasma samples can be stored for up to 28 days at 2-8ºC, so that they can be processed together in batches for the detection of IFNγ by ELISA, thus allowing an easy implementation of the assay in the laboratory workflow. Then, the IFNγ ELISA was performed in a Dynex DS2Ⓡ automated ELISA system. Final IFNγ values (IU/ml) for CD4+, CD4+&CD8+, and mitogen were obtained by subtracting the Nil value from the raw data. As cutoff has yet to be determined by the manufacturer, in an attempt to establish a cutoff value for the present study we added three standard deviation to the mean IFNγ value obtained from 25 unvaccinated and COVID-19 negative healthy donors (HD). Statistical analysis was performed with SPSS Statistics (version 26.0) and Statgraphics Centurion XVII. Comparison between groups was performed with the chi-square or Fisher's exact tests for categorical variables, and the Wilcoxon signed-rank and Mann-Whitney U tests for dependent and independent continuous variables, respectively. Figures were created with GraphPad Prism (version 9.02). A p-value <0.05 was considered statistically significant. A total of 50 participants were included. Their median age was 67 years (range, 19 to 97; 54% >60 years); 34 (68%) with (CoV2+) and 16 (32%) without (CoV2-) previous documented COVID- 19 . In the CoV2+ group, 94% were female compared to 75% in the CoV2-(p = 0.074). Differences in median age between the two cohorts were statistically significant (p = 0. Table 1 . At T0, 17/34 (50%) seropositive individuals were detected in the CoV2+ group, while 18/34 (53%) had at least one detectable QuantiFERON parameter of specific T cell response. Among the seropositive participants, four out of 17 did not reach positive levels in the QuantiFERON test; thus, five people had a negative anti-spike IgG result that had a detectable specific T cell response. The IFNγ-producing T cells were statistically higher among >60 years. There was no humoral nor T-cell response in the CoV2-group. Anti-spike IgG quantification after vaccination in CoV2-individuals showed statistically significant increases after each dose ( Figure 1A , Table 2 ). Eleven participants presented positivity at T1, but four (25%) did not reach levels above the assay cutoff. After the second dose (T2 and T3) all the CoV2-participants were seropositive, although one of them close to the cutoff. Regarding the cellular response, results for the naïve individuals showed a statistically significant increase after the first (T1) and the second doses (T2) in both T-cell subsets, and a month after the vaccination schedule ended, a tendency to decrease (p>0.05) was seen, also in both T-cell subsets (Figure 2A-B) . In 3 individuals, all aged >60 years, IFNγ was not detected in any of the lymphocyte populations studied (Table 1) , coinciding in T3 with the lowest antibodies values. Humoral (Table 2 ) and cellular responses (Table 3 ) in the CoV2-study were weaker in participants over 60 years compared to those younger, significantly so in most cases. In the previously-naturally-infected group (CoV2+), a significant increase was observed only after the first dose, in both anti-S IgGs and IFNγ-producing CD4+ and CD4+&CD8+ T-cells; and all median values in the different immune subsets were higher than in the CoV2-group. After the second dose, there was a slight increase (p<0.05) in antibody levels ( Figure 1C) , and, unlike the COV2-group, those over 60 years reached higher IgGs values at all-time points compared to those under 60 years ( Figure 1D ). The T response after the first dose (T1) was much greater than in the CoV2-group, and only in one individual was IFNγ not detected in the QuantiFERON assay. Two weeks after the second dose (T2), a significant decrease of IFNγ production by T cells was observed, followed by a subsequent increase in T3 to levels comparable to those obtained in T1 ( Figure 2C -D). All the CoV2+ participants showed QuantiFERON positive results at some time point, but at the end of the study, one individual <60 years was above the cutoff. When the cellular response was compared between CoV2+ age groups, all the values were significantly higher in the >60 year group (Table 3) . In the present study, we have observed two substantially different kinetics in the adaptive immune responses (humoral and cellular) to a 2-dose schedule mRNA vaccine among persons with and without previous COVID-19. All the individuals who had a proved COVID-19 infection approximately 10 months before experienced, after the first vaccine dose, a rapid and elevated increase in the production of anti-spike IgG levels, as observed in other recent studies [16] , [19] , requiring a 1:100 additional dilution of samples. This was accompanied by a potent activation of virus-specific CD4+ and CD8+ T effector memory cells, seen through the increase of IFNγ cytokine production, and that was previously detected in other studies of cellular immune response follow-up after COVID-19 infection [20] , [21] . In fact, what we have observed is a booster effect following the first vaccine dose, as the individuals were exposed to the same antigen twice [22] . Another study has shown a similar response after one dose of the same vaccine [19] . The second vaccine dose does not improved either humoral or cellular immune responses, as no significant increases in anti-spike IgG levels or specific IFNγproducing T cells was detected, as stated by others [16] . Additionally, we observed a significant decrease in IFNγ production by T cells early (two weeks) after the second vaccine dose, which may be related to a downregulation of the response, potentially related to regulatory T cells (Treg). Antiviral vaccination has been shown to stimulate Treg, which control exacerbated responses and prevent inflammatory damage, but that can limit the efficacy of the vaccines [23] , [24] . Improvements are being made with vaccine adjuvants in order to inhibit Treg, thus enhancing T cell response [25] , [26] . In any event, one month after the second dose IFNγ production by T cells increased over this previous time point (14 days after second dose), to levels similar to those achieved after the first dose, meaning that the downregulation could be transient, as suggested by others [25] . More investigation of that point is needed. Surprisingly, when we analysed results by age groups, we found that older people (>60 years) had more robust immune responses, displaying more than twice the serum antibody titres and IFNγ levels of CoV2+ people aged <60 years. This difference may be explained by a more exacerbated natural primary response in older people, as has been previously reported, that, in addition to implying more severity in this age-group [27] - [29] , also generates a greater T-cell memory production, allowing a more intense booster effect, even though the members of our studied cohort did not had severe COVID-19. In contrast to participants with previous COVID-19, individuals with no previous natural infection showed a slower and lower increase in both the humoral and cellular response markers. After the first dose, specific antibody levels and IFNγ production by T cells were strikingly poorer, remarkably so in older people, making the second dose essential. This may be explained by immunological senescence [30] that makes vaccination less effective in the elderly, as happens with other active immunisation processes. Even if the response may seem weak in the CoV2-group, more studies are assured in order to examine the time evolution of this immune subsets, considering also other markers that have not been studied here. In a recent study, an mRNA vaccine elicited a Th1-biased response, being the production of TNFα and IL-2 higher than IFNγ, which may in part explain why our results are not very high [31] . Also, it is important to note that the local immune response may differ from the peripheral blood compartment. Here, we demonstrate that a commercial IGRA, the QuantiFERON SARS-CoV-2 assay, is a valuable tool to analyse T cell response in large numbers of samples, being easy to implement in a routine laboratory workflow. To our knowledge, no other studies have been published using this specific assay. Similar approximation work has preceded ours [32] - [34] . We have proposed a cutoff for a specific T-cell response to SARS-CoV-2 of at least one result above 0.10 and 0.25 IU/ml for CD4+ and CD4+&CD8+ T cells, respectively. In a previous work, Murugesan et al. [32] developed an in-house IGRA assay for detection of SARS-CoV-2, and median IFNγ results in healthy donors was 0.01 (IQR 0-0.01) IU/ml, comparable to ours (0.00 IU/ml). They found that stimulation with the entire SARS-CoV-2 proteome showed higher results in COVID-19 convalescent people, which is not our case (the vaccination target is the S protein). Another study found a better IFNγ response after stimulation with a spike mega-pool compared to a remainder-antigens mega-pool [33] , probably because the latter did not include the S-region. Even so, for purposes other than vaccine response, it is important to note that T cell response to SARS-CoV-2 has been proven to bind a number of epitopes in the non-S region [11] . In our study, 5 out of 17 participants who had been previously infected with SARS-CoV-2 (29%) had a detectable T cell response by QuantiFERON with negative anti-spike results, demonstrating that the assay may be also useful in detecting seronegative individuals with previous infection, as suggested by others [10] , [12] . This aspect is of relevance because it can help to better estimate the exposed / immune population, and to evaluate the efficacy of the vaccines, taking into account the potential waning of antibody responses over time, and the fact that specific CD4+ and CD8+ T cells appear to confer broad and long-lasting protection to severe reinfection [10] . A SARS-CoV-2 IGRA test has been shown to anticipate the antibody response in some COVID-19 acute cases [34] , thus it may also be of interest in diagnosis. It is also notable that, like us, some published data did not detect a non-specific response by using an IGRA test [32] , [33] , contrary to what others have found with flow cytometry assays [10] , which may be related to the nature of the test. In fact, we detected some "residual response" in HD, which was higher for the IFNγ+CD4+&CD8+ T cell subset than for the IFNγ+CD4+ T cell one, which may also been explained by the different epitopes used to stimulate each subset, being RBD and S1/S2, respectively. It has been published by others [35] , that the CD4+ T cell reactivity to SARS-CoV-2 in HD was mainly due to the C-terminal S epitopes -corresponding to the S2 subunit-, which have a higher homology to the S proteins of other common human coronaviruses; in comparison to N-terminal epitopes -where the RBD is situated-. In order to detect specific T-cell response to SARS-CoV-2 vaccine, we raised the cutoff values of the tests above the highest value obtained from the uninfected donors. Another study detected IFNγ production by ELISpot [7] , showing that a 2-dose BNT162b2 schedule successfully primed a strong CD4+ T cell response (better than that of CD8+, in accordance with Murugesan et al. [32] ), which was higher for a two dose than a single schedule. According to our data, the CoV2-group benefits equally from the second dose. To sum up, even though more investigation is needed, the QuantiFERON assay could help in the future as a surrogate marker of protective immunity to SARS-CoV-2 or severe COVID-19. Our study is robust because we were able to assess samples from all participants at all interval times, permitting an optimal follow-up of immune response until one month after the second dose of the mRNA vaccine. However, there are limitations in the present work: the total number of participants in each group is low, a female bias exists in both CoV2-/Cov2+ groups, and there are statistically differences in the age between both groups (due to the characteristics of the studied cohort), thus limiting the generalisation of our results. Also, the studied immune subsets, the stimulating epitopes and the detected cytokine (IFNγ) may underrepresent the global response. Our work supports the strategy that a second vaccine dose is not a priority in people with previous COVID-19. This approach would make it possible to optimise the available doses of vaccines without increasing the risk of infection, allowing a faster expansion of vaccination coverage, which is now imperative. For this strategy to work, however, it is necessary to know with certainty who has previously had COVID-19. Thus, we recommend a pre-vaccination epidemiological survey and/or serological investigation in doubtful cases. Alternatively, SARS-CoV-2 serology and T-cell response could be scheduled after the first dose, to demonstrate the described booster effect and assist in deciding if a second dose is advisable. The implementation of more automated assays, such as ELISA and QuantiFERON, may help in generalising the proposed scheme. We thank to the staff of the Microbiology Department which have participated in the processing of samples, especially Silvia Navarro and Leticia Bueno. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. SARS-CoV-2 vaccines in development Primary exposure to SARS-CoV-2 protects against reinfection in rhesus macaques SARS-CoV-2 infection protects against rechallenge in rhesus macaques Neutralizing antibodies correlate with protection from SARS-CoV-2 in humans during a fishery vessel outbreak with a high attack rate Safety and Immunogenicity of Two RNA-Based Covid-19 Vaccine Candidates Evaluation of the mRNA-1273 Vaccine against SARS-CoV-2 in Nonhuman Primates COVID-19 vaccine BNT162b1 elicits human antibody and T H 1 T cell responses A decade after SARS: strategies for controlling emerging coronaviruses Balancing Expediency and Scientific Rigor in Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine Development Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19 Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals Intrafamilial exposure to SARS-CoV-2 associated with cellular immune response without Seroconversion, France Persistence of SARS-CoV-2 specific B-and T-cell responses in convalescent COVID-19 patients 6-8 months after the infection Persistence of SARS-CoV-2-specific antibodies in COVID-19 patients T cell and antibody responses induced by a single dose of ChAdOx1 nCoV-19 (AZD1222) vaccine in a phase 1/2 clinical trial Antibody Responses in Seropositive Persons after a Single Dose of SARS-CoV-2 mRNA Vaccine Evaluation of suitable target antigens and immunoassays for high-accuracy immune monitoring of cytomegalovirus and Epstein-Barr virus-specific T cells as targets of interest in immunotherapeutic approaches Interferon-γ release assays or tuberculin skin test for detection and management of latent tuberculosis infection: a systematic review and meta-analysis Effect of previous SARS-CoV-2 infection on humoral and T-cell responses to single-dose BNT162b2 vaccine Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection Functional SARS-CoV-2-Specific Immune Memory Persists after Mild COVID-19 Heterologous prime-boost vaccination Regulatory T cell frequencies and phenotypes following anti-viral vaccination Vaccine-induced antigen-specific regulatory T cells attenuate the antiviral immunity against acute influenza virus infection Regulatory T cell frequencies and phenotypes following anti-viral vaccination) and EFPIA companies' in kind contribution Regulatory T cells as adjuvant target for enhancing the viral disease vaccine efficacy Potential lethal outbreak of coronavirus disease (COVID-19) among the elderly in retirement homes and long-term facilities Articles Estimates of the severity of coronavirus disease 2019: a model-based analysis Comparison of commercial lateral flow immunoassays and ELISA for SARS-CoV-2 antibody detection Immunosenescence and herd immunity: With an ever-increasing aging population do we need to rethink vaccine schedules? An mRNA Vaccine against SARS-CoV-2 -Preliminary Report Interferon-gamma release assay for accurate detection of SARS-CoV-2 T cell response A whole blood test to measure SARS-CoV-2-specific response in COVID-19 patients Spike is the most recognized antigen in the whole-blood platform in both acute and convalescent COVID-19 patients SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19