Evaluation of the Immune Response Induced by CoronaVac 28-Day Schedule Vaccination in a Healthy Population Group

Alejandro Escobar, Felipe E Reyes-López, Mónica L Acevedo, Luis Alonso-Palomares, Fernando Valiente-Echeverría, Ricardo Soto-Rifo, Hugo Portillo, Jimena Gatica, Ivan Flores, Estefanía Nova-Lamperti, Carlos Barrera-Avalos, María Rosa Bono, Leonardo Vargas, Valeska Simon, Elias Leiva-Salcedo, Cecilia Vial, Juan Hormazabal, Lina Jimena Cortes, Daniel Valdés, Ana M Sandino, Mónica Imarai, Claudio Acuña-Castillo, Alejandro Escobar, Felipe E Reyes-López, Mónica L Acevedo, Luis Alonso-Palomares, Fernando Valiente-Echeverría, Ricardo Soto-Rifo, Hugo Portillo, Jimena Gatica, Ivan Flores, Estefanía Nova-Lamperti, Carlos Barrera-Avalos, María Rosa Bono, Leonardo Vargas, Valeska Simon, Elias Leiva-Salcedo, Cecilia Vial, Juan Hormazabal, Lina Jimena Cortes, Daniel Valdés, Ana M Sandino, Mónica Imarai, Claudio Acuña-Castillo

Abstract

CoronaVac vaccine from Sinovac Life Science is currently being used in several countries. In Chile, the effectiveness of preventing hospitalization is higher than 80% with a vaccination schedule. However, to date, there are no data about immune response induction or specific memory. For this reason, we recruited 15 volunteers without previous suspected/diagnosed COVID-19 and with negative PCR over time to evaluate the immune response to CoronaVac 28 and 90 days after the second immunization (dpi). The CoronaVac administration induces total and neutralizing anti-spike antibodies in all vaccinated volunteers at 28 and 90 dpi. Furthermore, using ELISpot analysis to assay cellular immune responses against SARS-CoV-2 spike protein, we found an increase in IFN-gamma- and Granzyme B-producing cells in vaccinated volunteers at 28 and 90 dpi. Together, our results indicate that CoronaVac induces a robust humoral immune response and cellular immune memory of at least 90 dpi.

Keywords: COVID-19; CoronaVac; SARS-CoV-2; herd immunity; immunological memory; neutralizing antibodies; vaccine.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Escobar, Reyes-López, Acevedo, Alonso-Palomares, Valiente-Echeverría, Soto-Rifo, Portillo, Gatica, Flores, Nova-Lamperti, Barrera-Avalos, Bono, Vargas, Simon, Leiva-Salcedo, Vial, Hormazabal, Cortes, Valdés, Sandino, Imarai and Acuña-Castillo.

Figures

Figure 1
Figure 1
Pro- and anti-inflammatory cytokines in the plasma from CoronaVac-vaccinated volunteers. The levels of (A) IL-8, (B) IL-12, (C) IL-6, (D) IL-1β, (E) IL-10, and (F) TNF-α were measured by flow cytometry as described in Materials and Methods. Data represented the cytokine level on each volunteer before (Day 0) and 28 days after the second immunization (dpi). The data show individual paired values. Asterisk (**) represents significant difference (p < 0.01). n/s indicates non-significant differences.
Figure 2
Figure 2
Total anti-S antibodies in the plasma of CoronaVac-vaccinated volunteers. Anti-S antibodies were quantified in plasma obtained prior to immunization (Day 0), and 28 and 90 days after the second immunization (dpi). (A) Antibody titers are expressed as the area under the ELISA OD curve (AUC) made from four serial dilutions (1/200, 1/400, 1/800, and 1/1600) for each volunteer (▾). The data show individual paired values. (B) Detail of the antibody titers for the volunteers with complete follow-up (n = 15) throughout the study (data from A;○), and those with no sample on some of the time points evaluated (•) were graphed and shown with median. Asterisk (*) represents statistically significant difference compared to baseline (p < 0.05). Hashtag (#) represents statistically significant difference between 28 dpi and 90 dpi samples. n/s, not significant (p < 0.05).
Figure 3
Figure 3
Total anti-S neutralizing antibodies in the plasma of CoronaVac-vaccinated volunteers. The neutralization curves from plasma samples of volunteers were obtained by using a pseudotyped viral particle (HIV-1–SΔ19). Samples were titrated in triplicate at serial threefold dilutions and expressed as percent of neutralization. (A) Total anti-S neutralizing antibodies before vaccination (Day 0). (B) Total anti-S neutralizing antibodies at 28 days after the second immunization (dpi). (C) Total anti-S neutralizing antibodies at 90 dpi (D) IC50 calculated based on inhibition rates and depicted for each sample obtained from volunteers with a complete follow-up schedule (□) (n = 15). The data show individual paired values. (E) IC50 calculated for the total data (n = 21 volunteers, including those with no complete follow-up schedule) were graphed and shown with median. Open circles (○): data from (D) Black circles (•): volunteers with no sample on some of the time-points evaluated. Asterisk (*) represents statistically significant difference obtained from the comparison against day 0 (p < 0.05). Hashtag (#) represents statistically significant difference between 28 dpi and 90 dpi samples. n/s, not significant (p < 0.05).
Figure 4
Figure 4
Cytokine secreting cells of vaccinated volunteers in vitro induced by SARS-CoV-2 S protein. (A) IFN-γ (○). (B) Granzyme (•). The number of ELISPOT-forming cells (SFC) per 3 × 105 was quantitated in PBMC, obtained before vaccination (indicated as "Day 0"). Twenty-eight and 90 days after the second immunization (dpi) (n = 15 volunteers), the data show individual paired values. (C) Total data from 21 volunteers (same volunteers from (A, B) and others with no sample on some of the time points evaluated) were graphed and shown with median for IFN-γ (○) and Granzyme (•). Asterisk (*) represents statistically significant difference (p < 0.05) obtained from the comparison against Day 0. n/s indicates non-significant differences.

References

    1. Tregoning JS, Flight KE, Higham SL, Wang Z, Pierce BF. Progress of the COVID-19 Vaccine Effort: Viruses, Vaccines and Variants Versus Efficacy, Effectiveness and Escape. Nat Rev Immunol (2021) 21(10):626–36. doi: 10.1038/s41577-021-00592-1
    1. Mulligan MJ, Lyke KE, Kitchin N, Absalon J, Gurtman A, Lockhart S, et al. . Phase I/II Study of COVID-19 RNA Vaccine BNT162b1 in Adults. Nature (2020) 586:589–93. doi: 10.1038/s41586-020-2639-4
    1. Palacios R, Patiño EG, de Oliveira Piorelli R, Conde MTRP, Batista AP, Zeng G, et al. . Double-Blind, Randomized, Placebo-Controlled Phase III Clinical Trial to Evaluate the Efficacy and Safety of Treating Healthcare Professionals With the Adsorbed COVID-19 (Inactivated) Vaccine Manufactured by Sinovac – PROFISCOV: A Structured Summary of a. Trials (2020) 21:21–3. doi: 10.1186/s13063-020-04775-4
    1. Bueno SM, Abarca K, González PA, Gálvez NMS, Soto JA, Duarte LF, et al. . Interim Report : Safety and Immunogenicity of an Inactivated Vaccine. medRxiv (2021). doi: 10.1101/2021.03.31.21254494
    1. Ministerio de Salud Gobierno de Chile . Ficha Vacuna Contra Sars-Cov-2 Vacuna Coronavac Laboratorio Sinovac Life Science® (2021). Available at: (Accessed August 23, 2021).
    1. Jara A, Undurraga EA, González C, Paredes F, Fontecilla T, Jara G, et al. . Effectiveness of an Inactivated SARS-CoV-2 Vaccine in Chile. N Engl J Med (2021) 385:875–84. doi: 10.1056/nejmoa2107715
    1. Huang AT, Garcia-Carreras B, Hitchings MDT, Yang B, Katzelnick LC, Rattigan SM, et al. . A Systematic Review of Antibody Mediated Immunity to Coronaviruses: Kinetics, Correlates of Protection, and Association With Severity. Nat Commun (2020) 11:1–16. doi: 10.1038/s41467-020-18450-4
    1. Beltrán-Pavez C, Riquelme-Barrios S, Oyarzún-Arrau A, Gaete-Argel A, González-Stegmaier R, Cereceda-Solis K, et al. . Insights Into Neutralizing Antibody Responses in Individuals Exposed to SARS-CoV-2 in Chile. Sci Adv (2021) 7(7):eabe6855. doi: 10.1126/sciadv.abe6855
    1. Palgen JL, Tchitchek N, Elhmouzi-Younes J, Delandre S, Namet I, Rosenbaum P, et al. . Prime and Boost Vaccination Elicit a Distinct Innate Myeloid Cell Immune Response. Sci Rep (2018) 8:1–18. doi: 10.1038/s41598-018-21222-2
    1. Channappanavar R, Zhao J. Perlman S. T Cell-Mediated Immune Response to Respiratory Coronaviruses. Immunol Res (2014) 59:118–28. doi: 10.1007/s12026-014-8534-z
    1. Dosch SF, Mahajan SD, Collins AR. SARS Coronavirus Spike Protein-Induced Innate Immune Response Occurs via Activation of the NF-κb Pathway in Human Monocyte Macrophages In Vitro . Virus Res (2009) 142:19–27. doi: 10.1016/j.virusres.2009.01.005
    1. Li T, Kenney AD, Liu H, Fiches GN, Zhou D, Ayan Biswas J, et al. . SARS-CoV-2 Nsp14 Activates NF-κb Signaling and Induces IL-8 Upregulation. BioRxiv (2021). doi: 10.1101/2021.05.26.445787
    1. Li J, Rong L, Cui R, Feng J, Jin Y, Yu Y, et al. . Dynamic Changes in Serum IL-6, IL-8, and IL-10 Are Associated With the Outcome of Patients With Severe COVID-19 in ICU. Res Sq (2021) 10(4):3706–14. doi: 10.21203/-83336/v1
    1. Amanna IJ, Carlson NE, Slifka MK. Duration of Humoral Immunity to Common Viral and Vaccine Antigens. N Engl J Med (2007) 357:1903–15. doi: 10.1542/peds.2008-2139LLLL
    1. Khoury DS, Cromer D, Reynaldi A, Schlub TE, Wheatley AK, Juno JA, et al. . Neutralizing Antibody Levels Are Highly Predictive of Immune Protection From Symptomatic SARS-CoV-2 Infection. Nat Med (2021) 27:1205–11. doi: 10.1038/s41591-021-01377-8
    1. Vabret N, Britton GJ, Gruber C, Hegde S, Kim J, Kuksin M, et al. . Immunology of COVID-19: Current State of the Science. Immunity (2020) 52:910–41. doi: 10.1016/j.immuni.2020.05.002
    1. Sahin U, Muik A, Derhovanessian E, Vogler I, Kranz LM, Vormehr M, et al. . COVID-19 Vaccine BNT162b1 Elicits Human Antibody and TH1 T Cell Responses. Nature (2020) 586:594–9. doi: 10.1038/s41586-020-2814-7
    1. Yang G, Feng F, Li X, Zhang T, Li X, Li B. Changes of Lymphocyte Subsets in Patients With COVID-19 and Clinical Significance: A Case-Control Observational Study. J Bio-X Res (2021) 4:36–9. doi: 10.1097/jbr.0000000000000089

Source: PubMed

3
Sottoscrivi