SARS-CoV-2 vaccine booster in solid organ transplant recipients previously immunised with inactivated versus mRNA vaccines: A prospective cohort study

Martín Dib, Nicole Le Corre, Catalina Ortiz, Daniel García, Marcela Ferrés, Constanza Martinez-Valdebenito, Cinthya Ruiz-Tagle, María José Ojeda, Manuel A Espinoza, Aquiles Jara, Juan Pablo Arab, Ricardo Rabagliati, Cecilia Vizcaya, María Elena Ceballos, Mauricio Sarmiento, Sebastián Mondaca, Macarena Viñuela, Antonia Pastore, Vania Szwarcfiter, Elizabeth Galdames, Aldo Barrera, Pablo Castro, Nicolás Ms Gálvez, Jorge A Soto, Susan M Bueno, Alexis M Kalergis, Bruno Nervi, M Elvira Balcells, Martín Dib, Nicole Le Corre, Catalina Ortiz, Daniel García, Marcela Ferrés, Constanza Martinez-Valdebenito, Cinthya Ruiz-Tagle, María José Ojeda, Manuel A Espinoza, Aquiles Jara, Juan Pablo Arab, Ricardo Rabagliati, Cecilia Vizcaya, María Elena Ceballos, Mauricio Sarmiento, Sebastián Mondaca, Macarena Viñuela, Antonia Pastore, Vania Szwarcfiter, Elizabeth Galdames, Aldo Barrera, Pablo Castro, Nicolás Ms Gálvez, Jorge A Soto, Susan M Bueno, Alexis M Kalergis, Bruno Nervi, M Elvira Balcells

Abstract

Background: Solid-organ transplant (SOT) recipients have worse COVID-19 outcomes than general population and effective immunisation in these patients is essential but more difficult to reach. We aimed to determine the immunogenicity of an mRNA SARS-CoV-2 vaccine booster in SOT recipients previously immunised with either inactivated or homologous SARS-CoV-2 mRNA vaccine.

Methods: Prospective cohort study of SOT recipients under medical care at Red de Salud UC-CHRISTUS, Chile, previously vaccinated with either CoronaVac or BNT162b2. All participants received a BNT162b2 vaccine booster. The primary study end point was anti-SARS-CoV-2 total IgG antibodies (TAb) seropositivity at 8-12 weeks (56-84 days) post booster. Secondary end points included neutralising antibodies (NAb) and specific T-cell responses.

Findings: A total of 140 (50% kidney, 38% liver, 6% heart) SOT recipients (mean age 54 [13.6] years; 64 [46%] women) were included. Of them, 62 had homologous (three doses of BNT162b2) and 78 heterologous vaccine schedules (two doses of CoronaVac followed by BNT162b2 booster). Boosters were received at a median of 21.3 weeks after primary vaccination. The proportion achieving TAb seropositivity (82.3% vs 65.4%, P = 0.035) and NAb positivity (77.4% vs 55.1%, P = 0.007) were higher for the homologous versus the heterologous group. On the other hand, the number of IFN-γ and IL-2 secreting SARS-CoV-2-specific T-cells did not differ significantly between groups.

Interpretation: This cohort study shows that homologous mRNA vaccine priming plus boosting in SOT recipients, reaches a significantly higher humoral immune response than inactivated SARS-CoV-2 vaccine priming followed by heterologous mRNA booster.

Funding: School of Medicine, UC-Chile and ANID.ClinicalTrials.gov ID: NCT05124509.

Keywords: BNT162b2; COVID-19; CoronaVac; SARS-CoV-2; Solid organ transplant; Vaccine; Vaccine immunogenicity.

Conflict of interest statement

S.M.B., N.L.C. and A.K. reported having participated as leading scientists for design of CoronaVac clinical trials sponsored in Chile by Pontificia Universidad Católica de Chile and in collaboration with Sinovac Biotech (NCT04651790 and NCT04992260).

© 2022 The Author(s).

Figures

Figure 1
Figure 1
Study flow-chart. Adult solid organ recipients transplanted within 10 years prior to study initiation, under medical care at the Transplant Institute, Red de Salud UC CHRISTUS.
Figure 2
Figure 2
Humoral response against SARS-CoV-2 in SOT recipients with an homologous or heterologous vaccination schedule. Serum was obtained between 8-12 weeks after the mRNA vaccine in the homologous group (n = 62) and the heterologous group (n = 78). Distribution for (a) frequency of total IgG (TAb) anti-S1 positivity (≥11 relative units per ml, RU/ml), (b) total IgG anti-S1 geometric mean concentration (GMC) (95%CI), RU/ml), (c) neutralising antibodies (NAb) positivity (≥30% of inhibition rate) and (d) neutralising activity (median (IQR) of percentage of inhibition). Dotted line in (b) and (d) show seropositivity cut-off. Statistical significance was calculated with Fisher test (a,c), Mann-Whitney (b,d); two-tailed P values are shown.
Figure 3
Figure 3
Evaluation of IFN-γ and IL-2 secreting Spot Forming T cells in SOT recipients with homologous and heterologous vaccination schedules. PBMCs (3×105 cells) obtained between 8-12 weeks after mRNA vaccine in the homologous group (n = 29) and the heterologous group (n = 31) were stimulated with megapool of peptides (MP S-R), or megapool of peptides (MP CD8) from SARS-CoV-2 proteins. (a) IFN-γ-secreting spot forming T cells (SFC) and (b) IL-2-secreting SFC were quantified by ELISPOT. Medians and interquartile ranges are shown. Statistical significance was calculated with Mann-Whitney test.

References

    1. Feikin DR, Higdon MM, Abu-Raddad LJ, et al. Duration of effectiveness of vaccines against SARS-CoV-2 infection and COVID-19 disease: results of a systematic review and meta-regression. Lancet. 2022;399(10328):924–944.
    1. Ministerio de Salud de Chile . 2021. Plan Nacional de Vacunacion Covid. Available from.
    1. Sauré D, O'Ryan M, Torres JP, Zuniga M, Santelices E, Basso LJ. Dynamic IgG seropositivity after rollout of CoronaVac and BNT162b2 COVID-19 vaccines in Chile: a sentinel surveillance study. Lancet Infect Dis. 2021;22(1):56–63.
    1. World Health Organization . 2021. Interim Statement on Booster Doses for COVID-19 Vaccination. Available from.
    1. Danziger-Isakov L, Blumberg EA, Manuel O, Sester M. Impact of COVID-19 in solid organ transplant recipients. Am J Transplant. 2021;21(3):925–937.
    1. Marion O, Del Bello A, Abravanel F, et al. Safety and immunogenicity of Anti-SARS-CoV-2 messenger RNA vaccines in recipients of solid organ transplants. Ann Intern Med. 2021;174(9):1336–1338.
    1. Balcells ME, Le Corre N, Durán J, et al. Reduced immune response to inactivated SARS-CoV-2 vaccine in a cohort of immunocompromised patients in Chile. Clin Infect Dis. 2022;75(1):e594–e602. doi: 10.1093/cid/ciac167.
    1. Caillard S, Chavarot N, Bertrand D, et al. Occurrence of severe COVID-19 in vaccinated transplant patients. Kidney Int. 2021;100(2):477–479.
    1. World Health Organization . 2021. Interim Recommendations for an Extended Primary Series with an Additional Vaccine Dose for COVID-19 Vaccination in Immunocompromised Persons. [updated Interim guidance, 26 October 2021. Available from.
    1. Grifoni A, Weiskopf D, Ramirez SI, 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) 1489-501.e15.
    1. Del Bello A, Abravanel F, Marion O, et al. Efficiency of a boost with a third dose of anti-SARS-CoV-2 messenger RNA-based vaccines in solid organ transplant recipients. Am J Transplant. 2022;22(1):322–323.
    1. Choi EM. COVID-19 vaccines for low- and middle-income countries. Trans R Soc Trop Med Hyg. 2021;115(5):447–456.
    1. Medina-Pestana J, Covas DT, Viana LA, et al. Inactivated whole-virus vaccine triggers low response against SARS-CoV-2 infection among renal transplant patients: prospective phase 4 study results. Transplantation. 2022;106(4):853–861.
    1. Heeger PS, Larsen CP, Segev DL. Implications of defective immune responses in SARS-CoV-2 vaccinated organ transplant recipients. Sci Immunol. 2021;6(61):eabj6513.
    1. Efros O, Anteby R, Halfon M, Meisel E, Klang E, Soffer S. Efficacy and safety of third dose of the COVID-19 vaccine among solid organ transplant recipients: a systemic review and meta-analysis. Vaccines. 2022;10(1):95.
    1. Kamar N, Abravanel F, Marion O, et al. Assessment of 4 doses of SARS-CoV-2 messenger RNA–based vaccine in recipients of a solid organ transplant. JAMA Netw Open. 2021;4(11) e2136030-e.
    1. Pozzetto B, Legros V, Djebali S, Barateau V, Guibert N, Villard M, et al. Immunogenicity and efficacy of heterologous ChAdOx1–BNT162b2 vaccination. Nature. 2021;600:701–706.
    1. (FDA) USFaDA . 2021. Coronavirus (COVID-19) Update: FDA Expands Eligibility for COVID-19 Vaccine Boosters. [updated November 19, 2021. Available from.
    1. Mok CKP, Chen C, Yiu K, et al. A randomized clinical trial using CoronaVac or BNT162b2 vaccine as a third dose in adults vaccinated with two doses of CoronaVac. Am J Respir Crit Care Med. 2022;205(7):844–847.
    1. Schmidt T, Klemis V, Schub D, et al. Cellular immunity predominates over humoral immunity after homologous and heterologous mRNA and vector-based COVID-19 vaccine regimens in solid organ transplant recipients. Am J Transplant. 2021;21(12):3990–4002.
    1. Reindl-Schwaighofer R, Heinzel A, Mayrdorfer M, et al. Comparison of SARS-CoV-2 antibody response 4 weeks after homologous vs heterologous third vaccine dose in kidney transplant recipients: a randomized clinical trial. JAMA Intern Med. 2021;182(2):165–171.
    1. Masset C, Ville S, Garandeau C, et al. Observations on improving COVID-19 vaccination responses in kidney transplant recipients: heterologous vaccination and immunosuppression modulation. Kidney Int. 2022;101(3):642–645.
    1. Ferreira VH, Marinelli T, Ierullo M, et al. Severe acute respiratory syndrome coronavirus 2 infection induces greater T-Cell responses compared to vaccination in solid organ transplant recipients. J Infect Dis. 2021;224(11):1849–1860.
    1. Cucchiari D, Egri N, Bodro M, et al. Cellular and humoral response after MRNA-1273 SARS-CoV-2 vaccine in kidney transplant recipients. Am J Transplant. 2021;21(8):2727–2739.
    1. Herrera S, Colmenero J, Pascal M, et al. Cellular and humoral immune response after mRNA-1273 SARS-CoV-2 vaccine in liver and heart transplant recipients. Am J Transplant. 2021;21(12):3971–3979.
    1. Dai Z, Konieczny BT, Lakkis FG. The dual role of IL-2 in the generation and maintenance of CD8+ memory T cells. J Immunol. 2000;165(6):3031–3036.
    1. Gao Y, Cai C, Grifoni A, et al. Ancestral SARS-CoV-2-specific T cells cross-recognize the Omicron variant. Nat Med. 2022;28(3):472–476.
    1. Aslam S, Danziger-Isakov L, Mehra MR. COVID-19 vaccination immune paresis in heart and lung transplantation. J Heart Lung Transplant. 2021;40(8):763–766.
    1. Caillard S, Thaunat O. COVID-19 vaccination in kidney transplant recipients. Nat Rev Nephrol. 2021;17(12):785–787.
    1. Visco-Comandini U, Castilletti C, Lionetti R, et al. High prevalence of asymptomatic SARS-CoV-2 infection in a cohort of liver transplant recipients in central Italy. J Liver Transplant. 2022;5

Source: PubMed

3
Se inscrever