Next Article in Journal
Renal Side Effects of COVID-19 Vaccination
Next Article in Special Issue
Utility of IL-6 in the Diagnosis, Treatment and Prognosis of COVID-19 Patients: A Longitudinal Study
Previous Article in Journal
Short-Term Adverse Effects Following Booster Dose of Inactivated-Virus vs. Adenoviral-Vector COVID-19 Vaccines in Algeria: A Cross-Sectional Study of the General Population
Previous Article in Special Issue
Nirmatrelvir/Ritonavir and Molnupiravir in the Treatment of Mild/Moderate COVID-19: Results of a Real-Life Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Cellular and Humoral SARS-CoV-2 Vaccination Responses in 192 Adult Recipients of Allogeneic Hematopoietic Cell Transplantation

1
Department of Medicine I/Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
2
Clinical Trials Unit, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
3
Institute of Virology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
*
Author to whom correspondence should be addressed.
Vaccines 2022, 10(11), 1782; https://doi.org/10.3390/vaccines10111782
Submission received: 22 September 2022 / Revised: 19 October 2022 / Accepted: 21 October 2022 / Published: 23 October 2022
(This article belongs to the Special Issue Global Trends of COVID-19 Vaccination and Immunology)

Abstract

:
To determine factors influencing the vaccination response against SARS-CoV-2 is of importance in recipients of allogeneic hematopoietic cell transplantation (allo-HCT) as they display an increased mortality after SARS-CoV-2 infection, an increased risk of extended viral persistence and reduced vaccination response. Real-life data on anti-SARS-CoV-2-S1-IgG titers (n = 192) and IFN-γ release (n = 110) of allo-HCT recipients were obtained using commercially available, validated assays after vaccination with either mRNA (Comirnaty™, Pfizer-BioNTech™, NY, US and Mainz, Germany or Spikevax™, Moderna™, Cambridge, Massachusetts, US) or vector-based vaccines (Vaxzevria™,AstraZeneca™, Cambridge, UK or Janssen COVID-19 vaccine™Johnson/Johnson, New Brunswick, New Jersey, US), or after a heterologous protocol (vector/mRNA). Humoral response (78% response rate) was influenced by age, time after transplantation, the usage of antithymocyte globulin (ATG) and ongoing immunosuppression, specifically corticosteroids. High counts of B cells during the vaccination period correlated with a humoral response. Only half (55%) of participants showed a cellular vaccination response. It depended on age, time after transplantation, ongoing immunosuppression with ciclosporin A, chronic graft-versus-host disease (cGvHD) and vaccination type, with vector-based protocols favoring a response. Cellular response failure correlated with a higher CD8+ count and activated/HLA-DR+ T cells one year after transplantation. Our data provide the basis to assess both humoral and cellular responses after SARS-CoV2 vaccination in daily practice, thereby opening up the possibility to identify patients at risk.

1. Introduction

Mortality after severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection in recipients of allogeneic hematopoietic cell transplantation (allo-HCT) ranged between 16–40% during the early phase of the pandemic, before the emergence of the omicron variant. Risk factors for SARS-CoV-2 mortality after allo-HCT have been reported to be older age, earlier time after allo-HCT, immunosuppression and male sex [1,2,3]. In healthy individuals, multiple studies have shown that the vaccination against SARS-CoV-2 protects against severe courses of infection [4]. In allo-HCT recipients, studies on SARS-CoV-2 infection and mortality after vaccination are scarce [5]. However, several studies have assessed anti-S1 IgG levels as a surrogate marker for vaccination effectiveness—in most cases, after the use of mRNA vaccines. Risk factors for humoral nonresponders after cellular therapy have slightly varied between cohorts, but B-cell depletion, lymphocyte counts <1000/μL, early vaccination after allo-HCT, ongoing immunosuppression and active graft-versus-host disease (GvHD) have been among the main factors affecting titers [6,7,8,9,10,11,12,13,14,15,16,17]. Although easy to obtain, the humoral immune response only reflects one aspect in adaptive immunity. It has been shown that T cells, especially CD8+, are mobilized early after mRNA vaccination [18] and SARS-CoV-2 infection elicits durable T-cell responses [19]. As there is no standardized routine assay to determine cellular vaccination response, data on T-cell-mediated immunity in allo-HCT recipients after vaccinations are still scarce and results are heterogeneous [9,20,21,22].
Here, we report real-life data on both humoral response and interferon-γ (IFN-γ) release after SARS-CoV-2 vaccination in a large cohort of allo-HCT recipients after different vaccination schemes (mRNA, vector vaccines and heterologous schemes with mRNA/vector). We assessed the impact of transplantation details, lymphocyte subpopulations and the differential effect of single immunosuppressive agents on vaccination responses.

2. Methods

2.1. Patients

This retrospective single-center study was performed at Freiburg University Medical Center after approval by the local Ethics Committee (EK-FR: 21-1590). We included 192 adult allo-HCT recipients with an assessment of humoral and/or cellular vaccination response on a routine basis and at least one SARS-CoV-2 vaccination. Data were assessed from December 2020 until October 2021.

2.2. Detection of SARS-CoV-2 Antibodies

Anti-SARS-CoV-S1-IgG was determined using a commercially available assay detecting IgG antibodies against S1-RBD (SARS-CoV-2 IgG, Siemens™, Munich, Germany). Anti-S1-IgG titers of >21.8 BAU/mL were classified as a humoral response.

2.3. SARS-CoV-2 IFN-γ Release Assay

The IFN-γ release assay (IGRA) was performed as described previously with a SARS-CoV2 IGRA (EUROIMMUN™, Lübeck, Germany). In brief, whole blood was incubated in stimulation tubes containing a peptide pool based on the S1 domain of the SARS-CoV-2 spike protein for 20–24 h with positive and negative stimulation controls. IFN-γ concentration was measured via an ELISA [23].

2.4. Lymphocyte Counts

Lymphocyte subsets were determined via flow cytometry after full blood staining on an FACS CANTO II™ and analyzed with FACS Diva™ (Becton Dickinson™, Franklin Lakes, New Jersey) at two time points: (1) The peri-vaccination period was defined as 0 to 3 months prior to the last or only documented vaccination date by month. (2) The time period of one year was defined as 330 to 390 days after allo-HCT. If there were multiple measurements during the defined time period, the average value was taken.

2.5. Statistical Analysis

Data were analyzed using R-Studio™ (Boston, Massachusetts, US) version 1.4.1717, Apache OpenOffice™ (Wakefield, Massachusetts) version 4.1.7. and Inkscape™ version 1.1.1. For continuous variables, normality was assessed visually. Group comparisons of data with a Gaussian distribution were performed using a two sided t-test, and non-normally distributed data were tested using a Wilcoxon rank-sum test. For categorical variables, a chi-squared test was used to assess differences. The associations between various parameters and the humoral and cellular response were analyzed using logistic regression models. The models were determined by including relevant factors based on univariate analysis and performing automated stepwise selection of independent variables using a combination of backward elimination and forward selection.

3. Results

3.1. Patient Cohort

We included 192 adult allo-HCT recipients (Table 1). Due to the data cut-off date (October 2021), the majority of patients received two vaccinations according to the recommendation of German vaccination guidelines. Patients were vaccinated predominately with mRNA-based vaccines (Comirnaty™, Pfizer-BioNTech™, NY, USA and Mainz, Germany or Spikevax™, Moderna™, Cambridge, MA, USA n = 129, 67%). A vector-based immunization (Vaxzevria™,AstraZeneca™, Cambridge, UK or Janssen COVID-19 vaccine™ Johnson/Johnson, New Brunswick, New Jersey,) was used in 10%, n = 20. A heterologous protocol of first using a vector-based agent, followed by a booster with an mRNA-based vaccination was used in 10 patients (5%). The heterologous protocol was implemented in Germany in spring 2021 for individuals below the age of 60 after the recognition of vaccine-induced immune thrombotic thrombocytopenia [24]. The time between allo-HCT and best vaccination response ranged between 4 months and 26 years with a median of 3.3 years for humoral and 3.4 years for cellular response. The majority of patients were off immunosuppressive therapy (n = 118, 61%) at the time of vaccination. For the 74 individuals (39%) with active immunosuppression, ruxolitinib was the agent most commonly used (n = 23), followed by ciclosporin A (n = 22) and prednisolone (n = 15).

3.2. Humoral Vaccination Response Depends on Age, Time after Allo-HCT, Antithymocyte Globulin (ATG) Usage and Active Immunosuppression

Anti-SARS-CoV-S1-IgG was assessed in all 192 patients. Of those, 150 patients (78%) responded, whereas 42 (22%) were nonresponders. Humoral responders were significantly younger compared with nonresponders, with a median age of 60 (IQR = 49.4–67.4) vs. 64.5 years (IQR = 55.3–70.8, p < 0.05). Responders were also younger at allo-HCT compared with nonresponders, with a median age at transplantation of 53.5 (IQR = 42.5–62.6) vs. 59.5 (IQR = 50.7–68.9) years (p < 0.01, Figure 1A). Additionally, the time from allo-HCT to humoral response was significantly longer in the responder group (median of 4 years, IQR = 1.7–7.7) vs. 1.3 years (IQR = 0.7–4.5, p < 0.01) in the nonresponder group (Figure 1A).
GvHD prophylaxis as well as ongoing immunosuppression differed in the two groups (Figure 1B). ATG-based GvHD prophylaxis was more prevalent in nonresponders (Figure 1B, p < 0.05). Among the humoral responders, 33% were on active immunosuppression at vaccination compared with 60% of the nonresponders (p < 0.05). Particularly, the use of prednisolone (8/15 responders, 53%, p < 0.01) and ciclosporin A (14/22 responders, 64%, p < 0.05) impaired specific antibody production compared to individuals without active immunosuppression (101/118 responders, 86%). The median daily prednisolone dosage was similar in humoral responders (7.5 mg) and nonresponders (10 mg). Of note, ruxolitinib did not seem to have an impact on humoral response (18/23 responders, 78%) compared to patients off active immunosuppression (86% responders, Figure 1B). Donor type and intensity of conditioning chemotherapy did not influence humoral response rates. Both groups predominantly received reduced toxicity myeloablative conditioning regimens (Figure 1B). Intriguingly, individuals receiving a heterologous vaccination scheme of a one-time vector followed by an mRNA-based vaccine showed a 100% humoral response rate (n = 10). However, compared to the response rate of mRNA- and vector-based vaccination of 78% (n = 129) and 75% (n = 20), respectively, this difference was not statistically significant (Figure 1B).
To consolidate the above results, we conducted a multivariate analysis to predict failure in the humoral response (Supplemental Table S1). In concordance to the univariate analysis, the variables determining the model were immunosuppression in general, the usage of prednisolone, the usage of ATG and age at transplantation, confirming the findings of the univariate analysis.

3.3. Cellular Vaccination Response Depends on Age, Ciclosporin a Usage, Vaccination Scheme and cGvHD

IFN-γ release after S1 peptide stimulation was determined to assess cellular vaccination response in 110 patients. Of those, 60 individuals (55%) responded, whereas 50 (45%) did not. In healthcare workers, the response rate after vaccination with the same assay has been 100% [23].
Similar to humoral response, in allo-HCT recipients the median age at vaccination (responders 55.5 years, IQR = 45.1–64.6; nonresponder 65.7 years, IQR = 59.7–70.6; p < 0.01) and at transplantation (responders 47.6, IQR = 61.0–32.0; nonresponder 62.1 years, IQR = 52.4–66.1; p < 0.01; Figure 1A,C) was lower in cellular responders compared with nonresponders. Additionally, the median time from transplantation to response was longer in cellular responders (4.0 years, IQR = 1.6–9.0) compared to nonresponders (median 2.1 years, IQR = 1.0–5.3; p = 0.057; Figure 1C).
The use of ATG-based GvHD prophylaxis was equally distributed among cellular responders and nonresponders (63% ATG usage in responders vs. 74% in nonresponders, p = 0.32). The influence of active immunosuppressive therapy was not as pronounced for cellular response as for humoral response. Among IGRA responders, 70% (n = 42) were off immunosuppressive therapy compared to 56% (n = 28) of nonresponders (p = 0.19). Among the different immunosuppressive agents, ciclosporin A had a significant impact on cellular response rates (4/14, 29% responders) compared with individuals off immunosuppression (42/70, 60% responders, p < 0.05). Patients treated with prednisolone or ruxolitinib had lower but not significantly different IGRA response rates compared to the non-immunosuppressed population (Figure 1D).
Among cellular responders, mismatch unrelated donors (MMUD) were more prevalent compared to nonresponders (MMUD cellular responder 18%, n = 11; nonresponder 8%, n = 4; p = 0.072). Matched related donors (MRD) were less prevalent among cellular responders (MRD cellular responder 17%, n = 10; nonresponder 36%, n = 18; Figure 1D).
Myeloablative conditioning (MAC) was used in 23% (n = 40) of responders and 6% of nonresponders (n = 3), possibly reflecting the younger age of MAC recipients. Additionally, reduced toxicity MAC regimens were used less frequently (55%, n = 33) in responders versus nonresponders (74%, n = 37; p < 0.05; Figure 1D).
With regard to the vaccination scheme, patients receiving a one-time vector followed by an mRNA-based vaccine had a 100% cellular response rate (n = 8). Patients vaccinated with only vector-based agents showed a response rate of 67% (n = 6) and patients immunized with only mRNA-based vaccines, a response rate of 48% (n = 75, p < 0.05, Figure 1D).
To consolidate the results of the univariate analyses, we used a multivariate analysis to predict cellular response failure. The variables determining the model were the vaccination scheme, age at transplantation, the use of ciclosporin A and if the patient at one time had moderate or severe cGvHD (Supplemental Table S2). For the regression model, patients vaccinated with only vector-based protocols and patients receiving the heterologous protocol were grouped due to the low number of patients in each group. The latter showed a significantly smaller likelihood for cellular response failure when compared to only mRNA-based vaccination (OR for nonresponse compared to mRNA 0.12, p = 0.01). cGvHD in our univariate analysis did not seem to influence cellular response (univariate 30% of responders vs. 38% of nonresponders with moderate or severe cGvHD, p = 0.50). In our bilinear logistic regression model, moderate or severe cGvHD increased the likelihood of cellular response failure upon vaccination (OR for non-response 2.52, p = 0.065).
In summary, age, vaccination scheme, cGvHD and use of ciclosporin A influenced IFN-γ release in allo-HCT recipients.

3.4. Influence of Lymphocyte Subsets on Humoral and Cellular Vaccination Response

To assess the influence of immunological reconstitution on humoral and cellular immunization responses, we analyzed lymphocyte subpopulations up to three months prior to vaccination (peri-vaccination period n = 78 for humoral, n = 49 for cellular response assessment, Supplemental Figure S1 left column). In patients vaccinated more than one year after allo-HCT, we assessed lymphocyte counts in vaccination responders and nonresponders one year after transplantation (Supplemental Figure S1 right column, n = 109 for humoral, n = 58 for cellular response assessment).
B-cell counts in the peri-vaccination period were higher in humoral responders compared with nonresponders (median B-cell count 182 cells/µL vs. 28 cells/µL, p < 0.05). A trend towards higher B-lymphocyte counts was also observed in cellular responders vs. nonresponders (median B-cell count 180 cells/µL vs. 73 cells/µL, p = 0.07). Additionally, peri-vaccine CD4+ T-cell counts were higher in the humoral responder group compared with nonresponders (median CD4+ T-cell count 330/µL vs. 211/µL, p = 0.07). In contrast, CD4+-lymphocyte counts were comparable between cellular responders and nonresponders (median CD4+ T-cell count 278/µL vs. 223/µL, p = 0.45). NK-cell counts were higher for humoral responders compared to nonresponders (median NK-cell count 196 cells/µL vs. 146 cells/µL, p = 0.05). This difference was not as pronounced for cellular responders vs. nonresponders (median NK-cell count 203 cells/µL vs. 166 cells/µL, p = 0.42).
At one year after allo-HCT, B-, CD4+ and NK-lymphocyte counts were comparable in humoral responders and nonresponders (median B-cell count: 246 cells/µL vs. 250 cells/µL; CD4+ T cell: 239/µL vs. 196/µL; NK cell: 207 cells/µL vs. 170 cells/µL). Interestingly, cellular responders had lower CD8+-lymphocyte counts at one year after transplantation compared with nonresponders (median CD8+: IGRA responder 364 cells/µL vs. IGRA nonresponder 678 cells/µL, p < 0.05). A trend towards higher CD8+-lymphocyte counts in cellular nonresponders was also observed in the peri-vaccination period (median CD8+: IGRA responder 309 cells/µL vs. nonresponder 442 cells/µL, p = 0.21). Cellular nonresponders also showed higher activated, HLA-DR+ T-cell counts one year after transplantation compared to responders (median HLA-DR+ T cells IGRA responder 58/µL vs. IGRA nonresponder 88/µL, p < 0.05).
In summary, humoral vaccination response is positively associated with B-, CD4+- and NK-cell counts in the peri-vaccination period, whereas higher CD8+-lymphocyte counts seemed to negatively influence IFN-γ release, especially one year after allo-HCT.

4. Discussion

In this study, we evaluated the humoral and cellular SARS-CoV-2 vaccination responses of 192 recipients of allo-HCT. Our real-world data show similar humoral response rates of about 80% in accordance to previous studies [16,25,26,27], but were lower compared to the 100% response rate reported in healthy individuals [23,27]. Besides age and time after allo-HCT, antibody titers were reduced upon active glucocorticoid and ciclosporin A usage and ATG-based GvHD prophylaxis, confirming previous reports [10,27]. As our cohort also includes the—to our knowledge—largest cohort of vaccination responses in allo-HCT recipients under ruxolitinib treatment, we were able to investigate the differential effect of JAK inhibition. Interestingly, in contrast to ciclosporin A and corticosteroids, antibody response was not affected by the use of ruxolitinib, thereby pointing towards possible tapering strategies in GvHD treatment. Although it has become evident that sterile immunity against SARS-CoV-2 cannot be achieved, many studies use antibody titers—determined in a variety of assays—as a surrogate marker for protection against SARS-CoV-2 in immunocompromised patients. However, the exact cut-offs for the protection against infection or a severe course of the disease for different variants of the virus have not been determined. Additionally, long-term persistence of B cells without titers has been reported [28]. Thus, humoral response can currently be only an orientation to guide vaccination and infection prevention strategies in immunocompromised persons.
Results of cellular vaccination responses in allo-HCT recipients are heterogeneous and not widely available. The methods used were ELISpot and/or intracellular cytokine staining and reported response rates have ranged from <30% [9,22] to high response rates of >75% [20,21]. We used an easy to perform, standardized and validated assay with a high specificity of 96.3–100% and sensitivity of 75.4–86.9% to detect past infection [23]. This test is easy to implement in routine diagnostics. We observed a response rate of 55% in allo-HCT recipients compared to a 100% response rate in healthcare workers [23], underlining the fact that the cellular immune response in patients after allo-HCT is severely compromised. IFN-γ release was especially reduced with increasing age due to immunosuppression with ciclosporin A and cGvHD. How cellular immunity affects protection against SARS-CoV-2 infection and/or against a severe course, specifically in recipients of allo-HCT, is still to be determined and beyond the scope of this study. However, concerning the preserved effectiveness of cellular response against new variants of SARS-CoV-2 [29], longitudinal studies evaluating protection and infection using the IGRA implemented in routine diagnostics seem feasible.
With regard to lymphocyte subsets, we were able to confirm previous observations, as our cohort humoral response correlated with higher B-, NK- and CD4-cell counts in the vaccination period [9,25,27,30,31]. Intriguingly, cellular response failure in our collective correlated with higher CD8+ and activated HLA-DR+ T cells one year after transplantation. Of note, the median time to best cellular response was 3.4 years in our cohort. Ram and colleagues reported a similar finding with a correlation between a recent low CD4+/CD8+ ratio for cellular response failure [9]. We hypothesize that higher activated CD8-cell counts are surrogate markers for impaired immune reconstitution after allo-HCT. Additionally, an inverted CD4/8 ratio has been reported in the context of immunosenescence in people living with human immunodeficiency virus (HIV) and as a marker of immune aging [32,33].
Due to the changing vaccination recommendations during our data acquisition, our collective contains the largest number of vector-based vaccinated allo-HCT recipients showing both high humoral and cellular response rates. Thus, in the collective of allo-HCT recipients, vector-based vaccination seems to provide superior immunization against SARS-CoV-2. This observation was also reported for the humoral response after the first vaccination [17]. Both mRNA- and vector-based vaccines are applied without adjuvants and elicit an immune response through intrinsic mechanisms. mRNA vaccines use the immune-activating properties of single- and double-stranded RNA via Toll-like receptors (TLR) and the inflammasome. Vector-based vaccines activate the immune system in both a TLR-dependent and -independent way [34,35]. We hypothesize that the broader immune activation with vector-based vaccines might be beneficial in immunocompromised hosts. Further studies will need to investigate if vector-based immunization protocols are more effective in preventing severe SARS-CoV-2 infection in allo-HCT recipients.
In summary, we provide a comprehensive analysis of humoral and cellular vaccination responses in a large cohort of adult allo-HCT recipients. The IGRA used in our manuscript can easily be included into routine practice in the future and provide a broader basis to guide decisions on vaccination strategies in this vulnerable patient cohort.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/vaccines10111782/s1, Figure S1: Lymphocyte subpopulations stratified according to vaccination response and timepoint title; Table S1: Multivariate analysis, humoral response failure; Table S2: Multivariate analysis, cellular response failure.

Author Contributions

T.M., D.H. and C.W. planned the study. D.H. provided vaccination responses. I.B. provided lymphocyte subsets. T.M. analyzed the data. G.I. provided statistical support. T.M. and C.W. extracted primary data and wrote the manuscript. R.W., H.B., J.F. and R.Z. oversaw the transplantation program and provided guidance and patient data. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Ethics Committee of University of Freiburg (EK-FR: 21-1590, 27 January 2022).

Informed Consent Statement

Informed consent was available from all subjects involved in the study.

Acknowledgments

We thank all patients for participation in this study. We thank Irmgard Matt for maintaining our database. We acknowledge support by the Open Access Publication Fund of the University of Freiburg.

Conflicts of Interest

The authors have nothing to disclose.

References

  1. Ljungman, P.; de la Camara, R.; Mikulska, M.; Tridello, G.; Aguado, B.; Zahrani, M.A.; Apperley, J.; Berceanu, A.; Bofarull, R.M.; Calbacho, M.; et al. COVID-19 and Stem Cell Transplantation; Results from an EBMT and GETH Multicenter Prospective Survey. Leukemia 2021, 35, 2885–2894. [Google Scholar] [CrossRef] [PubMed]
  2. Sharma, A.; Bhatt, N.S.; St Martin, A.; Abid, M.B.; Bloomquist, J.; Chemaly, R.F.; Dandoy, C.; Gauthier, J.; Gowda, L.; Perales, M.-A.; et al. Clinical Characteristics and Outcomes of COVID-19 in Haematopoietic Stem-Cell Transplantation Recipients: An Observational Cohort Study. Lancet Haematol. 2021, 8, e185–e193. [Google Scholar] [CrossRef]
  3. Schaffrath, J.; Brummer, C.; Wolff, D.; Holtick, U.; Kröger, N.; Bornhäuser, M.; Kraus, S.; Hilgendorf, I.; Blau, I.-W.; Penack, O.; et al. High Mortality of COVID-19 Early after Allogeneic Stem Cell Transplantation—A Retrospective Multicenter Analysis on Behalf of the German Cooperative Transplant Study Group. Transplant. Cell. Ther. 2022, 28, 337.e1–337.e10. [Google Scholar] [CrossRef]
  4. Ghazy, R.M.; Ashmawy, R.; Hamdy, N.A.; Elhadi, Y.A.M.; Reyad, O.A.; Elmalawany, D.; Almaghraby, A.; Shaaban, R.; Taha, S.H.N. Efficacy and Effectiveness of SARS-CoV-2 Vaccines: A Systematic Review and Meta-Analysis. Vaccines 2022, 10, 350. [Google Scholar] [CrossRef] [PubMed]
  5. Fendler, A.; Au, L.; Shepherd, S.T.C.; Byrne, F.; Cerrone, M.; Boos, L.A.; Rzeniewicz, K.; Gordon, W.; Shum, B.; Gerard, C.L.; et al. Functional Antibody and T Cell Immunity Following SARS-CoV-2 Infection, Including by Variants of Concern, in Patients with Cancer: The CAPTURE Study. Nat. Cancer 2021, 2, 1321–1337. [Google Scholar] [CrossRef]
  6. Jullien, M.; Le Bourgeois, A.; Coste-Burel, M.; Peterlin, P.; Garnier, A.; Rimbert, M.; Imbert, B.-M.; Le Gouill, S.; Moreau, P.; Mahe, B.; et al. B Cell Aplasia Is the Most Powerful Predictive Marker for Poor Humoral Response after BNT162b2 mRNA SARS-CoV-2 Vaccination in Recipients of Allogeneic Hematopoietic Stem Cell Transplantation. Transplant. Cell. Ther. 2022, 28, 279.e1. [Google Scholar] [CrossRef]
  7. Beerlage, A.; Leuzinger, K.; Valore, L.; Mathew, R.; Junker, T.; Drexler, B.; Passweg, J.R.; Hirsch, H.H.; Halter, J. Antibody Response to mRNA SARS-CoV-2 Vaccination in 182 Patients after Allogeneic Hematopoietic Cell Transplantation. Transpl. Infect. Dis. 2022, 24, e13828. [Google Scholar] [CrossRef]
  8. Chiarucci, M.; Paolasini, S.; Isidori, A.; Guiducci, B.; Loscocco, F.; Capalbo, M.; Visani, G. Immunological Response Against SARS-COV-2 After BNT162b2 Vaccine Administration Is Impaired in Allogeneic but Not in Autologous Stem Cell Transplant Recipients. Front. Oncol. 2021, 11, 737300. [Google Scholar] [CrossRef]
  9. Ram, R.; Hagin, D.; Kikozashvilli, N.; Freund, T.; Amit, O.; Bar-On, Y.; Beyar-Katz, O.; Shefer, G.; Moshiashvili, M.M.; Karni, C.; et al. Safety and Immunogenicity of the BNT162b2 mRNA COVID-19 Vaccine in Patients after Allogeneic HCT or CD19-Based CART Therapy-A Single-Center Prospective Cohort Study. Transplant. Cell. Ther. 2021, 27, 788–794. [Google Scholar] [CrossRef]
  10. Redjoul, R.; Le Bouter, A.; Beckerich, F.; Fourati, S.; Maury, S. Antibody Response after Second BNT162b2 Dose in Allogeneic HSCT Recipients. Lancet 2021, 398, 298–299. [Google Scholar] [CrossRef]
  11. Le Bourgeois, A.; Coste-Burel, M.; Guillaume, T.; Peterlin, P.; Garnier, A.; Béné, M.C.; Chevallier, P. Safety and Antibody Response After 1 and 2 Doses of BNT162b2 mRNA Vaccine in Recipients of Allogeneic Hematopoietic Stem Cell Transplant. JAMA Netw. Open 2021, 4, e2126344. [Google Scholar] [CrossRef] [PubMed]
  12. Piñana, J.L.; López-Corral, L.; Martino, R.; Montoro, J.; Vazquez, L.; Pérez, A.; Martin-Martin, G.; Facal-Malvar, A.; Ferrer, E.; Pascual, M.-J.; et al. SARS-CoV-2-Reactive Antibody Detection after SARS-CoV-2 Vaccination in Hematopoietic Stem Cell Transplant Recipients: Prospective Survey from the Spanish Hematopoietic Stem Cell Transplantation and Cell Therapy Group. Am. J. Hematol. 2022, 97, 30–42. [Google Scholar] [CrossRef] [PubMed]
  13. Maneikis, K.; Šablauskas, K.; Ringelevičiūtė, U.; Vaitekėnaitė, V.; Čekauskienė, R.; Kryžauskaitė, L.; Naumovas, D.; Banys, V.; Pečeliūnas, V.; Beinortas, T.; et al. Immunogenicity of the BNT162b2 COVID-19 mRNA Vaccine and Early Clinical Outcomes in Patients with Haematological Malignancies in Lithuania: A National Prospective Cohort Study. Lancet Haematol. 2021, 8, e583–e592. [Google Scholar] [CrossRef]
  14. Dhakal, B.; Abedin, S.; Fenske, T.; Chhabra, S.; Ledeboer, N.; Hari, P.; Hamadani, M. Response to SARS-CoV-2 Vaccination in Patients after Hematopoietic Cell Transplantation and CAR T-Cell Therapy. Blood 2021, 138, 1278–1281. [Google Scholar] [CrossRef] [PubMed]
  15. Ali, H.; Ngo, D.; Aribi, A.; Arslan, S.; Dadwal, S.; Marcucci, G.; Nakamura, R.; Forman, S.J.; Chen, J.; Al Malki, M.M. Safety and Tolerability of SARS-CoV2 Emergency-Use Authorized Vaccines for Allogeneic Hematopoietic Stem Cell Transplant Recipients. Transplant. Cell. Ther. 2021, 27, 938.e1–e938.e6. [Google Scholar] [CrossRef]
  16. Mamez, A.-C.; Pradier, A.; Giannotti, F.; Petitpas, A.; Urdiola, M.F.; Vu, D.-L.; Masouridi-Levrat, S.; Morin, S.; Dantin, C.; Clerc-Renaud, D.; et al. Antibody Responses to SARS-CoV2 Vaccination in Allogeneic Hematopoietic Stem Cell Transplant Recipients. Bone Marrow Transplant. 2021, 56, 3094–3096. [Google Scholar] [CrossRef] [PubMed]
  17. Easdale, S.; Shea, R.; Ellis, L.; Bazin, J.; Davis, K.; Dallas, F.; Thistlethwayte, E.; Ethell, M.; Potter, M.; Arias, C.; et al. Serologic Responses Following a Single Dose of SARS-Cov-2 Vaccination in Allogeneic Stem Cell Transplantation Recipients. Transplant. Cell. Ther. 2021, 27, 880.e1–e880.e4. [Google Scholar] [CrossRef]
  18. Oberhardt, V.; Luxenburger, H.; Kemming, J.; Schulien, I.; Ciminski, K.; Giese, S.; Csernalabics, B.; Lang-Meli, J.; Janowska, I.; Staniek, J.; et al. Rapid and Stable Mobilization of CD8+ T Cells by SARS-CoV-2 mRNA Vaccine. Nature 2021, 597, 268–273. [Google Scholar] [CrossRef]
  19. Zuo, J.; Dowell, A.C.; Pearce, H.; Verma, K.; Long, H.M.; Begum, J.; Aiano, F.; Amin-Chowdhury, Z.; Hoschler, K.; Brooks, T.; et al. Robust SARS-CoV-2-Specific T Cell Immunity Is Maintained at 6 Months Following Primary Infection. Nat. Immunol. 2021, 22, 620–626. [Google Scholar] [CrossRef]
  20. Clémenceau, B.; Guillaume, T.; Coste-Burel, M.; Peterlin, P.; Garnier, A.; Le Bourgeois, A.; Jullien, M.; Ollier, J.; Grain, A.; Béné, M.C.; et al. SARS-CoV-2 T-Cell Responses in Allogeneic Hematopoietic Stem Cell Recipients Following Two Doses of BNT162b2 mRNA Vaccine. Vaccines 2022, 10, 448. [Google Scholar] [CrossRef]
  21. Harrington, P.; Doores, K.J.; Saha, C.; Saunders, J.; Child, F.; Dillon, R.; Saglam, S.; Raj, K.; McLornan, D.; Avenoso, D.; et al. Repeated Vaccination against SARS-CoV-2 Elicits Robust Polyfunctional T Cell Response in Allogeneic Stem Cell Transplantation Recipients. Cancer Cell 2021, 39, 1654. [Google Scholar] [CrossRef] [PubMed]
  22. Lindemann, M.; Klisanin, V.; Thümmler, L.; Fisenkci, N.; Tsachakis-Mück, N.; Ditschkowski, M.; Schwarzkopf, S.; Klump, H.; Reinhardt, H.C.; Horn, P.A.; et al. Humoral and Cellular Vaccination Responses against SARS-CoV-2 in Hematopoietic Stem Cell Transplant Recipients. Vaccines 2021, 9, 1075. [Google Scholar] [CrossRef] [PubMed]
  23. Huzly, D.; Panning, M.; Smely, F.; Enders, M.; Komp, J.; Falcone, V.; Steinmann, D. Accuracy and Real Life Performance of a Novel Interferon-γ Release Assay for the Detection of SARS-CoV2 Specific T Cell Response. J. Clin. Virol. 2022, 148, 105098. [Google Scholar] [CrossRef] [PubMed]
  24. Greinacher, A.; Thiele, T.; Warkentin, T.E.; Weisser, K.; Kyrle, P.A.; Eichinger, S. Thrombotic Thrombocytopenia after ChAdOx1 nCov-19 Vaccination. N. Engl. J. Med. 2021, 384, 2092–2101. [Google Scholar] [CrossRef] [PubMed]
  25. Canti, L.; Humblet-Baron, S.; Desombere, I.; Neumann, J.; Pannus, P.; Heyndrickx, L.; Henry, A.; Servais, S.; Willems, E.; Ehx, G.; et al. Predictors of Neutralizing Antibody Response to BNT162b2 Vaccination in Allogeneic Hematopoietic Stem Cell Transplant Recipients. J. Hematol. Oncol. 2021, 14, 174. [Google Scholar] [CrossRef]
  26. Shem-Tov, N.; Yerushalmi, R.; Danylesko, I.; Litachevsky, V.; Levy, I.; Olmer, L.; Lusitg, Y.; Avigdor, A.; Nagler, A.; Shimoni, A.; et al. Immunogenicity and Safety of the BNT162b2 mRNA COVID-19 Vaccine in Haematopoietic Stem Cell Transplantation Recipients. Br. J. Haematol. 2022, 196, 884–891. [Google Scholar] [CrossRef]
  27. Tamari, R.; Politikos, I.; Knorr, D.A.; Vardhana, S.A.; Young, J.C.; Marcello, L.T.; Doddi, S.; Devlin, S.M.; Ramanathan, L.V.; Pessin, M.S.; et al. Predictors of Humoral Response to SARS-CoV-2 Vaccination after Hematopoietic Cell Transplantation and CAR T-Cell Therapy. Blood Cancer Discov. 2021, 2, 577–585. [Google Scholar] [CrossRef]
  28. Piano Mortari, E.; Russo, C.; Vinci, M.R.; Terreri, S.; Fernandez Salinas, A.; Piccioni, L.; Alteri, C.; Colagrossi, L.; Coltella, L.; Ranno, S.; et al. Highly Specific Memory B Cells Generation after the 2nd Dose of BNT162b2 Vaccine Compensate for the Decline of Serum Antibodies and Absence of Mucosal IgA. Cells 2021, 10, 2541. [Google Scholar] [CrossRef]
  29. Keeton, R.; Tincho, M.B.; Ngomti, A.; Baguma, R.; Benede, N.; Suzuki, A.; Khan, K.; Cele, S.; Bernstein, M.; Karim, F.; et al. T Cell Responses to SARS-CoV-2 Spike Cross-Recognize Omicron. Nature 2022, 603, 488–492. [Google Scholar] [CrossRef]
  30. Huang, A.; Cicin-Sain, C.; Pasin, C.; Epp, S.; Audigé, A.; Müller, N.J.; Nilsson, J.; Bankova, A.; Wolfensberger, N.; Vilinovszki, O.; et al. Antibody Response to SARS-CoV-2 Vaccination in Patients Following Allogeneic Hematopoietic Cell Transplantation. Transplant. Cell. Ther. 2022, 28, 214.e1–e214.e11. [Google Scholar] [CrossRef]
  31. Cuapio, A.; Boulouis, C.; Filipovic, I.; Wullimann, D.; Kammann, T.; Parrot, T.; Chen, P.; Akber, M.; Gao, Y.; Hammer, Q.; et al. NK Cell Frequencies, Function and Correlates to Vaccine Outcome in BNT162b2 mRNA Anti-SARS-CoV-2 Vaccinated Healthy and Immunocompromised Individuals. Mol. Med. 2022, 28, 20. [Google Scholar] [CrossRef] [PubMed]
  32. Serrano-Villar, S.; Sainz, T.; Lee, S.A.; Hunt, P.W.; Sinclair, E.; Shacklett, B.L.; Ferre, A.L.; Hayes, T.L.; Somsouk, M.; Hsue, P.Y.; et al. HIV-Infected Individuals with Low CD4/CD8 Ratio despite Effective Antiretroviral Therapy Exhibit Altered T Cell Subsets, Heightened CD8+ T Cell Activation, and Increased Risk of Non-AIDS Morbidity and Mortality. PLoS Pathog. 2014, 10, e1004078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Wikby, A.; Ferguson, F.; Forsey, R.; Thompson, J.; Strindhall, J.; Löfgren, S.; Nilsson, B.-O.; Ernerudh, J.; Pawelec, G.; Johansson, B. An Immune Risk Phenotype, Cognitive Impairment, and Survival in Very Late Life: Impact of Allostatic Load in Swedish Octogenarian and Nonagenarian Humans. J. Gerontol. A Biol. Sci. Med. Sci. 2005, 60, 556–565. [Google Scholar] [CrossRef] [PubMed]
  34. Zhu, J.; Huang, X.; Yang, Y. Innate Immune Response to Adenoviral Vectors Is Mediated by Both Toll-like Receptor-Dependent and -Independent Pathways. J. Virol. 2007, 81, 3170–3180. [Google Scholar] [CrossRef] [Green Version]
  35. Teijaro, J.R.; Farber, D.L. COVID-19 Vaccines: Modes of Immune Activation and Future Challenges. Nat. Rev. Immunol. 2021, 21, 195–197. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Humoral and cellular SARS-CoV-2 vaccination response. (A) Box plots display age at allo-HCT and the time from allo-HCT to best measured humoral (A) and cellular response (C) stratified according to responders and nonresponders. Points represent individual patients. (B,D) In the left column, pie charts show frequencies of usage of antithymocyte globulin (ATG), active ongoing treatment with immunosuppressants (ISP) and the usage of specific immunosuppressive agents in responders versus nonresponders. The right column displays frequencies of donor type, intensity of the conditioning chemotherapy and the vaccination protocol used in responders and nonresponders. Matched unrelated donors (MUD), matched related donors (MRD), mismatched unrelated donors (MMUD), mismatched related donors (MMRD), HLA-haploidentical donors (HAPLO), myeloablative conditioning (MAC), reduced toxicity myeloablative conditioning (RTC) and reduced intensity conditioning (RIC). Vaccination protocols displayed are based on only messenger RNA (mRNA) vaccines, only viral vector vaccines (V) or a heterologous combination of one-time viral vector vaccine followed by an mRNA vaccine (V + mRNA). *, p < 0.05.
Figure 1. Humoral and cellular SARS-CoV-2 vaccination response. (A) Box plots display age at allo-HCT and the time from allo-HCT to best measured humoral (A) and cellular response (C) stratified according to responders and nonresponders. Points represent individual patients. (B,D) In the left column, pie charts show frequencies of usage of antithymocyte globulin (ATG), active ongoing treatment with immunosuppressants (ISP) and the usage of specific immunosuppressive agents in responders versus nonresponders. The right column displays frequencies of donor type, intensity of the conditioning chemotherapy and the vaccination protocol used in responders and nonresponders. Matched unrelated donors (MUD), matched related donors (MRD), mismatched unrelated donors (MMUD), mismatched related donors (MMRD), HLA-haploidentical donors (HAPLO), myeloablative conditioning (MAC), reduced toxicity myeloablative conditioning (RTC) and reduced intensity conditioning (RIC). Vaccination protocols displayed are based on only messenger RNA (mRNA) vaccines, only viral vector vaccines (V) or a heterologous combination of one-time viral vector vaccine followed by an mRNA vaccine (V + mRNA). *, p < 0.05.
Vaccines 10 01782 g001
Table 1. Patient characteristics.
Table 1. Patient characteristics.
CharacteristicsEntire Cohort (n = 192)
Age at data cut-off (year), median (range)61 (21–83)
Age at allo-HCT (year), median (range)54 (19–79)
Time from allo-HCT to best humoral response (years), median (range)3.3 (0.35–26.03)
Time from allo-HCT to best cellular response (years), median (range)3.4 (0.41–26.03)
Male, n (%)115 (60)
Female, n (%)77 (40)
Number of vaccinations documented, n (%)
1192 (100)
2177 (92)
34 (2)
Type of vaccination, n (%)
mRNA129 (67)
Vector20 (10)
1 mRNA + 1 Vector10 (5)
Unconventionally vaccinated2 (1)
Not documented31 (16)
Baseline disease, n (%)
AML78 (41)
ALL10 (5)
MDS21 (11)
MDS/MPN6 (3)
Myeloma21 (11)
Other NHL20 (10)
Immunodeficiency5 (3)
Other7 (4)
Donor type, n (%)
MUD102 (53)
MRD52 (27)
MMUD29 (15)
MMRD2 (1)
HAPLO7 (4)
Graft source, n (%)
PBSC183 (95)
BM9 (5)
Conditioning intensity
MAC33 (17)
RTC119 (62)
RIC40 (21)
CMV, n (%)
−/−83 (43)
−/+36 (19)
+/−23 (12)
+/+50 (26)
GvHD, n (%)
aGvHD122 (64)
aGvHD Grade II or more76 (40)
cGvHD111 (58)
cGvHD moderate or severe74 (39)
Active immunosuppression, n (%)
None118 (61)
Ruxolitinib23 (12)
Ciclosporin A22 (11)
Prednisolone15 (8)
Prednisolone dose (mg), median (range)10 (2.50–50)
Everolimus6 (3)
Mycophenolic acid2 (1)
Other29 (15)
Combination of 2 agents11 (6)
Combination of 3 agents6 (3)
Time from vaccination to response (years)
For humoral response n = 167
median (IQR)0.15 (0.07–0.28)
For cellular response n = 113
median (IQR)0.18 (0.10–0.28)
Legend Table 1: allo-HCT—allogeneic hematopoietic cell transplantation, AML—acute myeloid leukemia, ALL—acute lymphatic leukemia, MDS—myelodysplastic syndrome, MPN—myeloproliferative neoplasia, NHL—non-Hodgkin lymphoma, (M)MUD— (mis)matched unrelated donor, (M)MRD— (mis)matched related donor, HAPLO—haploidentical donor, PBSC—peripheral blood stem cell, BM—bone marrow, MAC—myeloablative, RTC—reduced toxicity MAC, RIC—reduced intensity, CMV—cytomegalovirus, (a/c)GvHD—(acute/chronic) graft-versus-host disease.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Meyer, T.; Ihorst, G.; Bartsch, I.; Zeiser, R.; Wäsch, R.; Bertz, H.; Finke, J.; Huzly, D.; Wehr, C. Cellular and Humoral SARS-CoV-2 Vaccination Responses in 192 Adult Recipients of Allogeneic Hematopoietic Cell Transplantation. Vaccines 2022, 10, 1782. https://doi.org/10.3390/vaccines10111782

AMA Style

Meyer T, Ihorst G, Bartsch I, Zeiser R, Wäsch R, Bertz H, Finke J, Huzly D, Wehr C. Cellular and Humoral SARS-CoV-2 Vaccination Responses in 192 Adult Recipients of Allogeneic Hematopoietic Cell Transplantation. Vaccines. 2022; 10(11):1782. https://doi.org/10.3390/vaccines10111782

Chicago/Turabian Style

Meyer, Thomas, Gabriele Ihorst, Ingrid Bartsch, Robert Zeiser, Ralph Wäsch, Hartmut Bertz, Jürgen Finke, Daniela Huzly, and Claudia Wehr. 2022. "Cellular and Humoral SARS-CoV-2 Vaccination Responses in 192 Adult Recipients of Allogeneic Hematopoietic Cell Transplantation" Vaccines 10, no. 11: 1782. https://doi.org/10.3390/vaccines10111782

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop