Skip to main content
Erschienen in: Journal of Clinical Immunology 5/2023

Open Access 17.03.2023 | Original Article

Impaired B Cell Recall Memory and Reduced Antibody Avidity but Robust T Cell Response in CVID Patients After COVID-19 Vaccination

verfasst von: Sophie Steiner, Tatjana Schwarz, Victor M. Corman, Lara M. Jeworowski, Sandra Bauer, Christian Drosten, Carmen Scheibenbogen, Leif G. Hanitsch

Erschienen in: Journal of Clinical Immunology | Ausgabe 5/2023

Abstract

Purpose

Humoral and cellular immune responses were described after COVID-19 vaccination in patients with common variable immunodeficiency disorder (CVID). This study aimed to investigate SARS-CoV-2-specific antibody quality and memory function of B cell immunity as well as T cell responses after COVID-19 vaccination in seroresponding and non-responding CVID patients.

Methods

We evaluated antibody avidity and applied a memory B cell ELSPOT assay for functional B cell recall memory response to SARS-CoV-2 after COVID-19 vaccination in CVID seroresponders. We comparatively analyzed SARS-CoV-2 spike reactive polyfunctional T cell response and reactive peripheral follicular T helper cells (pTFH) by flow cytometry in seroresponding and non-seroresponding CVID patients. All CVID patients had previously failed to mount a humoral response to pneumococcal conjugate vaccine.

Results

SARS-CoV-2 spike antibody avidity of seroresponding CVID patients was significantly lower than in healthy controls. Only 30% of seroresponding CVID patients showed a minimal memory B cell recall response in ELISPOT assay. One hundred percent of CVID seroresponders and 83% of non-seroresponders had a detectable polyfunctional T cell response. Induction of antigen-specific CD4+CD154+CD137+CXCR5+ pTFH cells by the COVID-19 vaccine was higher in CVID seroresponder than in non-seroresponder. Levels of pTFH did not correlate with antibody response or avidity.

Conclusion

Reduced avidity and significantly impaired recall memory formation after COVID-19 vaccination in seroresponding CVID patients stress the importance of a more differentiated analysis of humoral immune response in CVID patients. Our observations challenge the clinical implications that follow the binary categorization into seroresponder and non-seroresponder.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10875-023-01468-w.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Coronavirus disease (COVID-19) is caused by severe respiratory syndrome coronavirus-2 (SARS-CoV-2), and has caused more than 500 million infections and over 6 million deaths worldwide since its emerging in late 2019 [1].
Patients with common variable immunodeficiency disorder (CVID), the most frequent clinically relevant primary immunodeficiency, are at higher risk for COVID-19-associated hospitalization and mortality [2] as well as for increased risk of prolonged or recurrent (breakthrough) SARS-CoV-2 infections [3]. Vaccination is considered to be the most effective and safest prophylactic measure in patients with primary immunodeficiency including genetically defined inborn errors of immunity (IEI) [4]. Cumulative data on COVID-19 vaccine response from more than 1500 patients with IEI have been reported with CVID being the most frequent underlying immunodeficiency [5]. Findings on humoral immune response after COVID-19 vaccination are variable with frequencies of seroresponding CVID patients ranging from 20 to 95% [612]. A positive T cellular immune response was reported in 46–83% of CVID patients [8, 1015]. Reasons for the observed variability might include differences in methodology and different vaccination regimens as well as clinical, immunological, and genetic heterogeneity of CVID patients.
More importantly, the detection of specific antibodies after COVID-19 vaccination raises important questions on the quality and longevity of the humoral immune response, since the seroconversion state has potential clinical implications for treatment in SARS-CoV-2-infected CVID patients. Functional assessments of humoral immunity already revealed that CVID patients express lower neutralizing antibody levels than healthy individuals [811, 15]. Avidity of generated antibodies was analyzed in two recent studies, reporting similar levels of antibody avidity in CVID patients and healthy individuals 4 weeks after second COVID-19 vaccination but without significant increase after more than two vaccinations [9, 16]. Detailed characterizations regarding B cell memory formation are limited and suggest an atypical memory formation [17]. Without affecting seroconversion rates, booster vaccination in CVID patients was shown to further increase antibody levels in some seroresponder [9, 18], while effects of boosting on specific T cell immunity is variable [9, 10].
In the present study, we evaluated antibody avidity and functional B cell recall memory responses to SARS-CoV-2 vaccination in CVID seroresponders. In addition, specific polyfunctional T cell response and the generation of SARS-CoV-2 specific follicular T helper cells (TFH) were assessed by flow cytometry in CVID seroresponder and non-seroresponder. All included CVID patients had a previously documented impaired specific antibody response to conjugated pneumococcal vaccination.

Methods

Study Subjects

Samples of 16 CVID patients before first and after the second SARS-CoV-2 vaccination were collected from the outpatient clinic for immunodeficiencies at the Institute for Medical Immunology, Charité Universitätsmedizin Berlin. All CVID patients were adults and diagnosed according to the criteria defined by the European Society for Immune Deficiency (ESID) [19]. CVID patients had an impaired vaccine response to pneumococcal conjugate vaccine (specific IgG antibodies below protective levels or low specific antibodies without increase after vaccination). Samples of 8 healthy controls (HC) before and after the second dose of SARS-CoV-2 vaccination were collected from laboratory employees at Charité Universitätsmedizin Berlin. All samples were collected between June and October 2021. During this period, SARS-CoV-2 B.1.617.2 (Delta) was the most predominant strain in Germany. All CVID patients and HC were infection naïve with no clinical history of SARS-CoV-2 infection, expressing negative spike antibodies before first vaccination (Table 2) and remaining seronegative for nucleocapsid (NP) after vaccination, to address possible infections between sampling time points (Supplementary Table S2).

Sample Preparation

Serum and heparinized whole blood was collected at a median of 133 days for CVID seroresponder (IQR: 24) and 128 days for CVID non-seroresponder (IQR: 34) after second COVID-19 vaccination. Samples from healthy individuals were collected at 32 days (IQR: 5) after second COVID-19 vaccination. Peripheral blood mononuclear cells (PBMCs) were isolated by density gradient centrifugation over Pancoll (PAN-Biotech, Germany) using Leucosep tubes (Greiner Bio-One). PBMCs were cryopreserved and stored in liquid nitrogen.

SARS-CoV-2 Antibody Serology

SARS-CoV-2 spike serum IgG against the S1 domain was assessed by ELISA according to the manufacturer’s instructions (Euroimmun Medizinische Labordiagnostika AG, Lübeck, Germany) using fully automated Euroimmun Analyzer I (Euroimmun Medizinische Labordiagnostika AG, Lübeck, Germany). To confirm results obtained by ELISA, a microarray-based multiparametric immunoassay for detection of IgG antibodies against SARS-CoV-2 spike and NP (SeraSpot® Anti-SARS-CoV-2 IgG, Seramun Diagnostica GmbH, Heidesee, Germany) was applied.

IgG Avidity Assay

To measure avidity of SARS-CoV-2 spike IgG antibodies, serum samples were analyzed by a modified SARS-CoV-2-S1 ELISA (Euroimmun) [20]. Serum samples were diluted 1:101 with sample buffer and incubated on plates pre-coated with recombinant SARS-CoV-2 spike (S1) proteins. After incubation for 1 h at 37 °C, wells were washed and 200 µL urea (5.5 M.) or 200 µL phosphate-buffered saline (PBS) was added to the plates and incubated for 10 min at 37 °C. After a washing step, conjugate and substrate were added according to the manufacturer’s instructions. OD was detected at 450 nm, and the relative avidity index was calculated by dividing the observed OD of the urea-treated sample by that of the PBS-treated sample, multiplied by 100 [20].

SARS-CoV-2 Interferon-Gamma Release Assay (IGRA)

IGRA (Euroimmun) for quantitative IFNγ release by SARS-CoV-2-specific T cells following second dose SARS-CoV-2 vaccine was performed according to the manufacturer’s instructions. In summary, 500 µl heparinized whole blood was added to three stimulation tubes coated with specific SARS-CoV-2 S1 peptide pool, mitogen control, and uncoated blank, respectively. Blood was incubated for 24 h at 37 °C, 5% CO2. Collected plasma was stored at − 20 °C until analysis by Quan-T-Cell ELISA (Euroimmun). According to manufacturer values ≥ 200 IU/ml are positive, values between 100 and 200 IU/ml are considered borderline.

T Cell Phenotyping for SARS-CoV-2 Spike Reactive T Cells by Flow Cytometry

For each experimental approach, patient and control samples before and after vaccination were simultaneously assessed. Cryopreserved PBMCs were thawed and rested for 24 h in IMDM/10% FCS/1% P/S at 37 °C, 5% CO2. Stimulation was performed with 1 µg/ml of SARS-CoV-2 S peptide pools for N- and C-terminal domains (PM-WCPV-S-1, JPT Peptide Technologies GmbH, Berlin). Superantigen staphylococcal enterotoxin B (SEB) was used (3 µg/ml) as positive and DMSO as background control. Secretion inhibitor brefeldin A (BFA) (15 µg/ml) was added to each condition after 2 h. Stimulation continued for 16 h. Cells were washed and extracellular markers for anti-human CCR7 AF488, CD45RA PE-Cy7, and Live/Dead Fixable Blue stained for 30 min at 37 °C, 5% CO2. After repeated washing, fixation/permeabilization buffer was applied (FoxP3 transcription factor staining buffer set, eBioscience) and incubated for 30 min at 4 °C. Intracellular staining was performed for anti-human CD3 BV650, CD4 PerCp-Cy5.5, CD8 BV510, CD137 PE, CD154 BV421, IL-2 APC, IFNγ BV605, TNFα AF700, and CXCR5 PE-Dazzle (Supplementary Table S3) for 30 min at 4 °C. CytoflexLX flow cytometer and FlowJo software version 10.6.2 were used for analysis. Unspecific activation was excluded by subtracting the background signal (DMSO only) from the peptide and SEB activated samples. A positive T cell response was defined as CD154+CD137+CD4+ T cells > 0.005% within total CD4+ T cell population and 20% above the background. Boolean combination gating was used for analysis of single and polyfunctional cytokine producing T cell subsets.

Memory B Cell ELISPOT Assay

Cryopreserved PBMCs were thawed and seeded in a 6-well plate at a concentration of 4 × 106 cells in 3 ml RPMI/10% FCS/1% P/S (culture medium) per well in the presence of 5% CO2 at 37 °C. B cell proliferation was induced by the protocol from Crotty et al. [21] with 6 µg/ml CpG, 100 ng/ml of Pokeweed mitogen (PWM), staphylococcus aureus Cowan (SAC) (1:10 000), and 50 μM β-Mercaptoethanol for 7 days (called SAC protocol hereafter).
For the detection of antibody secreting cells (ASC) in HC and seroresponsive CVID patients after the second dose of SARS-CoV-2 vaccine, ELISPOT assay (enzyme-linked immuno spot assay) was performed with expanded cells after the 7-day cell culture. 96-well MultiScreen Filter Plates (Merck Millipore) were coated overnight (ON) at 4 °C with 1 µg/ml stabilized trimeric spike protein SARS-CoV-2 (wild type Excell Gene), as well as 1.2 μg/ml goat anti-human IgG (Jackson ImmunoResearch), serving as positive control. PBS was applied as negative control to exclude unspecific antibody binding.
Expanded cells were plated in the 96-well MultiScreen Filter Plates in duplicate at dilutions of 2.5 × 105/100 µl (IgG) and 1 × 106/100 µl for SARS-CoV-2 S protein, 6.25 × 103/100 µl for IgG positive controls and incubated for 6 h. Wells were then thoroughly washed six times with PBS supplemented with 1% bovine serum albumin (BSA) and 0.05% Tween. 100 μl/well, goat anti-human IgG-HRP (1:500) (Invitrogen) secondary antibody was applied and incubated ON at 4 °C. Afterwards, wells were washed three times with PBS. Substrate buffer (0.3 M sodium acetat solution, 0.2 M acetic acid solution, Auqa dest., pH = 5.0), 3-amino-9-ethyl-carbazole (AEC)-dimethylformamide (DMF) solution (1:30) and 3% H2O2 (1:100)) was applied to reach spot development. AID ELISPOT reader and AID ELISPOT 7.0 iSpot software were used for analysis. Results were manually verified to exclude artificial spots and multiple counting.

B Cell Phenotyping by Flow Cytometry

FACS analysis of B cell subsets was performed on PBMCs of day 0 (ex vivo) and on day 7 after cell culture (in vitro). Cells were incubated with a LIVE/DEAD fixable Aqua (Thermo Fisher) for 30 min at RT. Extracellular staining with fluorescently conjugated antibodies CD3 PB, CD19 PE-Cy7, CD21 PE, CD24 PerCp-Cy5.5, CD27 FITC, CD38 AF700, IgM APC, and IgD APC-Cy7 (Supplementary Table S4) for 30 min at 4 °C was performed. CytoflexLX flow cytometer and FlowJo software version 10.6.2 were used for analysis. Gating for B cell phenotyping was performed according to EUROClass classification (Supplementary Table S5, Supplementary Fig. S1) [22].

Statistical Analyses

Unpaired comparisons across multiple groups were performed using the Kruskal–Wallis test with Dunn’s post-test for multiple comparisons to find significant differences among multiple investigated groups. If a significance was detected, two-tailed Mann–Whitney U test was performed for unpaired comparisons across two groups. In order to analyze responses before and after vaccination, the Wilcoxon matched pairs signed-rank test was applied for paired comparisons within a group. Correlation analyses were performed using Spearman’s rank correlation coefficient. Continuous variables are shown as median and interquartile range (IQR). A p-value of < 0.05 was considered statistically significant. GraphPad Prism version 9.3.1 was used for statistical analyses.

Results

Study Cohort Characteristics

Ten seroresponding (CVID R), 6 non-responding (CVID NR) CVID patients, and 8 healthy controls (HC) were analyzed unvaccinated and after second COVID-19 vaccination. Seroresponse was defined according to manufactures instruction by a ratio of  > 1,1 for spike-specific SARS-CoV-2 IgG antibody response in Euroimmun ELISA (Table 1). Data from SeraSpot analysis confirmed humoral immune response to spike (S1, S full, and RBD) in CVID R (Supplementary Table S2). Basic immunological parameters included IgG, IgA, IgM, CD3+, CD4+, CD8+, and CD19+ cells as well as NK cells and T and B cell subsets (see Table 2) and showed no significant differences between the CVID groups. In addition, clinical characterization included non-infectious (immune cytopenia, autoimmunity, lympho-proliferation, granulomatous lymphocytic interstitial lung disease) and infectious manifestations (recurrent pneumonia and bronchiectasis) which showed no significant differences (see Table 3). Age and gender were similar in CVID R and CVID NR (median age 57 years).
Table 1
Serological data of anti-SARS-CoV-2 spike IgG in CVID patients and healthy controls (EUROIMMUN ELISA©)
 
Before COVID-19 vaccination
After COVID-19 vaccination
IgG OD ratio
Result
IgG OD ratio
Result
CVID R 1
0.13
Negative
6.48
Reactive
CVID R 2
0.16
Negative
3.86
Reactive
CVID R 3
0.13
Negative
2.88
Reactive
CVID R 4
0.1
Negative
1.94
Reactive
CVID R 5
0.14
Negative
2.82
Reactive
CVID R 6
0.12
Negative
3.21
Reactive
CVID R 7
0.14
Negative
7.75
Reactive
CVID R 8
0.14
Negative
3.56
Reactive
CVID R 9
0.9
Negative
3.04
Reactive
CVID R 10
0.28
Negative
2.24
Reactive
CVID NR 1
0.14
Negative
0.14
Negative
CVID NR 2
0.15
Negative
0.66
Negative
CVID NR 3
0.14
Negative
0.28
Negative
CVID NR 4
0.25
Negative
0.23
Negative
CVID NR 5
0.14
Negative
0.48
Negative
CVID NR 6
0.23
Negative
0.15
Negative
HC-1
0.16
Negative
7.67
Reactive
HC-2
0.12
Negative
7.4
Reactive
HC-3
0.12
Negative
6.42
Reactive
HC-4
0.19
Negative
6.85
Reactive
HC-5
0.08
Negative
7.36
Reactive
HC-6
0.09
Negative
7.22
Reactive
HC-7
0.09
Negative
8.12
Reactive
HC-8
0.08
Negative
8.3
Reactive
COVID-19, coronavirus disease 2019; CVID, common variable immunodeficiency disorder; HC, healthy control; NR, non-seroresponder; OD, optical density; R, seroresponder
Table 2
Description of immunological parameter before COVID-19 vaccination comparing CVID seroresponder with CVID patients that failed to mount specific antibodies after completed COVID-19 vaccination
 
IgG in g/L prior to IgRT
IgA in g/L
IgM in g/L
CD4+ (cell count per nl)
CD8+ (cell count per nl)
CD19+ (cell count per nl)
NK (cell count per nl)
Naïve CD4+ CD45RA+ in % of CD4+
Naïve Bc in % of CD19+
MZ-like Bc in % of CD19+
IgM + MBC in % of CD19+
CS MBC in % of CD19+
Transitional Bc in % of CD19+
Activated Bc in % of CD19+
CS PB in % of CD19+
Normal range
7–16
0.7–4.0
0.4–2.3
0.5–1.2
0.3–0.8
0.1–0.4
0.1–0.4
 > 15
42.6–82.3
7.4–32.5
6.5–29.1
0.6–3.4
0.9–7.6
0.4–3.6
CVID R 1
1.30
0.06
0.11
0.59
0.72
0.96
0.39
10
71.9
4.3
0.3
0.5
8.7
12.6
0
CVID R 2
2.66
 < 0.1
0.06
0.41
0.14
0.14
0.09
34
67.3
14.5
1.4
0.5
2.4
13.2
0.1
CVID R 3
 < 0.3
0.06
0.05
0.57
0.61
0.28
0.09
21
86.2
5.7
0.3
1
4
1.6
0
CVID R 4
 < 0.3
 < 0.1
0.05
0.41
0.36
0.22
0.28
10
81.3
12.2
0.3
0.6
3.5
1.5
0
CVID R 5
2.00
0.06
0.1
0.55
0.23
0.07
0.06
6
71.4
3.5
0.3
1.3
11.4
12.3
0.1
CVID R 6
1.86
0.14
0.15
0.37
1.23
0.22
0.13
24
76.9
8.4
0.6
2.7
1.6
7.7
0.3
CVID R 7
3.70
0.59
0.4
0.69
0.34
0.24
0.07
62
79.8
7.6
0.4
0.8
1.7
9.1
0.1
CVID R 8
3.00
0.04
0.16
0.55
0.33
0.37
0.13
27
56.51
31.73
2.36
1.78
2.47
6.06
0.89
CVID R 9
0.77
 < 0.1
0.05
0.26
0.76
0.14
0.41
6
53.7
9
3.8
0.7
9.2
21.6
0.8
CVID R 10
1.69
0.06
0.28
0.33
0.5
0.12
0.07
15
64.4
12.9
1.9
1.6
0.5
18.7
0
Median values CVID R 1–10
1.78
0.06
0.13
0.48
0.43
0.22
0.11
18
71.65
8.7
0.5
0.9
2.985
10.7
0.1
CVID NR 1
1.66
0.06
0.15
0.32
0.33
0.19
0.1
21
76
3.5
2.5
0.2
6.6
3.3
0
CVID NR 2
3.60
0.25
0.18
0.43
0.23
0.06
0.02
5
70.9
4.7
0
1.3
8
14.7
0.5
CVID NR 3
0.33
0.06
0.05
0.5
0.28
0.16
0.26
21
79.3
11.2
0.6
0.8
1.6
4.8
0.4
CVID NR 4
 < 0.30
0.06
0.05
0.64
0.62
0.04
0.05
47
81.4
7.3
1.1
0.3
4.2
4.1
0
CVID NR 5
 < 0.3
 < 0.1
2.2
0.84
0.44
0.38
0.11
60
59.3
33.8
0
0.6
0.7
5.3
0.1
CVID NR 6
 < 0.3
 < 0.1
0.05
0.93
0.55
0.08
0.05
2
67.5
8.8
2.6
1.1
1.8
15.8
0
Median values CVID NR 1–6
0.32
0.08
0.1
0.57
0.385
0.12
0.08
21
73.45
8.05
0.85
0.7
3
5.05
0.05
p-value*
CVID R vs. CVID NR
0.21
0.52
0.94
0.30
0.54
0.21
0.17
0.94
0.87
0.74
0.99
0.28
0.73
0.56
0.80
Values are in bold to highlight that this line is a summary of the group (CVID R and CVID NR respectively)
Bc, B cell; CS, class-switched; CVID, common variable immunodeficiency disorder; g/L, grams per liter; HC, healthy control; IgRT, immunoglobulin replacement therapy; MBC, memory B cells; MZ, marginal zone; nl, nanoliters; NK, natural killer cells; NR, non-seroresponder; OD, optical density; PB, plasmablasts; R, seroresponder; bolded values = median; p-value calculated using Mann–Whitney U test
Table 3
Clinical characterization of CVID seroresponder (CVID R) and CVID non-seroresponder (CVID NR) before COVID-19 vaccination in % of affected patients
 
Immune cytopenia
Autoimmunity
Splenomegaly/lympho-proliferation
GLILD
Bronchiectasis
Rec. pneumonias
Immuno-suppression
Genetic diagnostics
CVID R
10%
20%
60%
10%
0%
30%
0
Negative in all patients
CVID NR
33%
0%
100%
0%
33%
50%
0
Negative in all patients
p-value
0.52
0.5
0.23
1
0.125
0.61
1
 
CVID, common variable immunodeficiency disorder; GLILD, granulomatous lymphocytic interstitial lung disease; NR, non-seroresponder; R, seroresponder; rec., recurrent

Impaired SARS-CoV-2 Spike Antibody Response in CVID Patients

Spike-specific SARS-CoV-2 IgG antibody response was analyzed in two different systems (ELISA and SeraSpot). ELISA indicated a significant increase of spike-specific SARS-CoV-2 IgG antibodies after COVID-19 vaccination in HC (IgG: p = 0.008) and CVID R patients (IgG: p = 0.002), but antibody levels were significantly lower in CVID R patients compared to HC (IgG: p = 0.002) (Fig. 1A). Data from SeraSpot analysis indicated negativity for NP, which is not induced by the spike-based COVID-19 vaccination but by a previous SARS-CoV-2 infection (Supplementary Table S2).

SARS-CoV-2 Spike Antibody Avidity Is Significantly Diminished in Seroresponding CVID Patients

SARS-CoV-2 spike antibody avidity of CVID R patients was significantly lower than in HC (p < 0,001; Fig. 1B). Avidity correlated with levels of SARS-CoV-2 spike IgG in HC (p = 0.01; r = 0.833, Fig. 1C) but not in CVID R patients (p = 0.2; r = 0.45 Fig. 1D).

Formation of B Cell Memory Is Impaired in CVID Patients Despite the Presence of Circulating Antibodies

It is unknown whether seroconversion in CVID patients could also result in the development of a functional B cell memory. Here, we aimed to study the functional memory B cell (MBC) response after COVID-19 vaccination in CVID R patients and HC. ELISPOT results were analyzed in combination with B cell subsets (for gating see Supplementary Fig. S1). For comparability, data was calculated per B cell and PB proportion within CVID patients and HC. IgG secreting cells, detected in the ELISPOT assay, were calculated per 10,000 PBs and 10,0000 MBC which were used on day 0 for in vitro stimulation.

Memory B Cell and Plasmablast Phenotype in Seropositive CVID Patients and HC

Based on flow cytometry staining at day 0 ex vivo and day 7 after in vitro stimulation using the SAC protocol, a decrease in both percentage and count of class-switched (CS) MBC was observed in HC (Fig. 2A). As expected, CVID R patients had initial significantly lower levels of CS MBC compared to HC (p < 0.0001). After expansion, CVID R patients elicited again significantly lower levels of CS MBC compared to HC (p = 0.003, Fig. 2A). CVID R patients also had significantly lower frequencies of CS PB ex vivo (p < 0.0001), and CS PB after in vitro stimulation (p < 0.0001, Fig. 2B). The decrease in frequency of MBC and increase in PB in HC indicates successful differentiation after in vitro stimulation. MBC of CVID R patients were also able to differentiate into PB following SAC stimulation, but to a much lower extent.

Deficient Memory B Cell Recall Response in Seropositive CVID Patients After COVID-19 Vaccination

Only 3/10 CVID R patients showed a minimal SARS-CoV-2 MBC recall response in the ELISPOT assay, whereas all HC showed a response (see Fig. 2C for exemplarily ELISPOT wells). CVID R patients had significantly lower SARS-CoV-2 ASCs per 10,000 CS MBC for S specific IgG (p = 0.0007) compared to HC (Fig. 2D). Regarding CS PB, ELISPOT elucidated a lower MBC recall response for S IgG (p = 0.001) in CVID R compared to HC as well (Fig. 2F). Whole IgG positive controls elicited high levels of ASCs in HC. Of note, whole IgG control responses were as well detectable in 7/10 CVID R patients. After normalizing spots per 10,000 CS PB and 10,000 CS MBC, counts did not differ significantly between the groups (p = 0.1 and p = 0.4 respectively, Fig. 2E and G).

SARS-CoV-2 T Cellular Immune Response Is Robustly Induced in CVID Patients After COVID-19 Vaccination

In addition to humoral immune responses, the T cellular response was examined to evaluate if a SARS-CoV-2-specific T cell response is induced as a result of COVID-19 vaccination. Seroresponsive, non-seroresponsive CVID patients and HCs were comparatively analyzed.

Quantitative IFNγ Release by SARS-CoV-2-Specific T Cells

The IGRA enables the quantitative determination of IFNγ release by SARS-CoV-2 T cells after pathogen-specific stimulation. IGRA revealed a positive response in 6 CVID R and 3 CVID NR patients. One patient of each group was borderline positive. Negative results were obtained from 3 CVID R and 2 CVID NR patients. Stimulation with PMA served as positive control and showed positive results in all CVID patients despite one of the seroresponding individuals, which in contrast had a positive SARS-CoV-2-specific response (Table 4).
Table 4
Post COVID-19 vaccination IFNγ-release assay of SARS-CoV-2 peptide and PMA stimulated whole blood in CVID seroresponder and CVID non-seroresponder
ID
SARS-CoV-2
IFNγ [mIU/ml]
Mitogen control PMA
IFNγ [mIU/ml]
CVID R 1
79.80
2403.20
CVID R 2
64.32
511.42
CVID R 3
147.15
2396.39
CVID R 4
677.59
2488.77
CVID R 5
227.29
2421.58
CVID R 6
618.80
44.74
CVID R 7
2483.27
2483.27
CVID R 8
202.30
2484.13
CVID R 9
2435.33
2435.33
CVID R 10
23.88
860.71
Median CVID R 1–10
214.78
2412.39
CVID NR 1
56.20
358.26
CVID NR 2
124.73
2306.53
CVID NR 3
466.27
2499.50
CVID NR 4
2499.50
2499.50
CVID NR 5
1128.41
938.55
CVID NR 6
34.58
2308.46
Median CVID NR 1–6
295.50
2307.50
CVID, common variable immunodeficiency disorder; mIU/ml, milli-international units per milliliter; NR, non-seroresponder; R, seroresponder; rec., recurrent, PMA, Phorbol-12-myristat-13-acetat

SARS-CoV-2 Spike Reactive Polyfunctional T Cell Responses

SARS-CoV-2 S peptide–activated T cell subsets were assessed by flow cytometry. Antigen-specific CD4+ T cells were investigated using activation markers CD137 and CD154 along with expression of cytokines IFNγ, TNFα, and IL-2. An activated T cell response was defined as > 0.005% of total CD4+ T cells and 20% above the background signal. Polyfunctional cytokine subsets were obtained by Boolean combination gating. Moreover, formation of TFH cells was investigated by staining of CXCR5 in activated CD4+ T cells (for gating, see Supplementary Fig. S2).
SARS-CoV-2 S reactive CD4+CD154+CD137+ T cells were induced in all HCs (N-term p = 0.016; C-term p = 0.008) and CVID R (N-term p = 0.004) after COVID-19 vaccination. In the group of CVID NR patients, 5 of 6 were able to generate a CD4+ T cell response to a similar extent than the two other groups (Fig. 3A). SEB positive control revealed comparable levels of CD4+CD154+CD137+ T cells before and after vaccination among all groups investigated indicating an intact T cell response. CVID NR patients showed slightly higher levels of activated CD4+ T cells post vaccination after SEB stimulation compared to CVID R patients (p = 0.02) (Fig. 3B).
Vaccination in our infection naïve study cohort induced comparable frequencies of polyfunctional activated CD4+CD154+CD137+ T cells in all groups. IFNγ+TNFα+IL-2+ triple-positive (tp) SARS-CoV-2 S N- and C-terminal reactive T cells significantly increased post vaccination in HC and CVID R (N-term: HC p = 0.008, CVID R p = 0.002; C-term: HC p = 0.008, CVID R p = 0.04). In the group of CVID NR, tp-activated CD4+ T cells were induced in 5/6 patients. Post vaccination tp cytokine responses did not differ between the three groups (Fig. 3C).

COVID-19 Vaccination Induces Spike-Specific Circulating TFH Cells

Higher frequencies of CD4+CD45RACXCR5+ peripheral TFH cells were observed in CVID patients than in HC (Fig. 3D; for gating, see Supplementary Fig. S2). Induction of antigen reactive CD4+CD154+CD137+CXCR5+ peripheral TFH cells by the COVID-19 vaccine was detected in all three groups (Fig. 3E). Frequencies of TFH cells significantly increased in response to stimulation with SARS-CoV-2 S N-terminal peptide pool in HC (p = 0.008) and CVID R (p = 0.02). In CVID NR, 5/6 patients showed an increase but did not reach statistical significance. Regarding stimulation with the C-terminal peptide, pool frequencies were higher in HC after vaccination (p = 0.008). Moreover, SARS-CoV-2 S C-terminal reactive pTFH were higher in HC compared to CVID NR (p = 0.008) as well as in CVID R compared to CVID NR (p = 0.02). Moreover, levels of SARS-CoV-2 spike–specific pTFH cells did not correlate with antibody levels, antibody avidity or frequency of SARS-CoV-2 spike reactive polyfunctional CD4+ cells (see Supplementary Fig. S3).

Discussion

Evaluation of humoral COVID-19 vaccine response revealed variable and in part surprisingly high rates of seroresponders among CVID patients. However, in a disease, which is defined by impaired antibody and B cell memory formation, quality and longevity of humoral immune response need to be considered.
In the present study, SARS-CoV-2 seroresponding and non-responding CVID patients as well as HC were comparatively analyzed for their antibody avidity and for the functional longevity of their humoral immune response by using a SARS-CoV-2 spike–specific MBC ELISPOT assay. In addition, T cellular immune response, including flow cytometric detection of SARS-CoV-2 reactive polyfunctional CD4+ T cells and TFH cells, was assessed.
Despite detectable SARS-CoV-2 antibodies after two COVID-19 vaccinations, humoral immune response in CVID patients differed substantially from healthy individuals as SARS-CoV-2 spike antibodies in CVID patients showed a significantly reduced avidity in comparison to HC. Our study complements recently published data on avidity in CVID patients after COVID-19 vaccination [9, 16]. In contrast to our data, Sauerwein et al. observed similar levels of specific SARS-CoV-2 antibody levels and avidity in HC and CVID patients after two vaccinations, but reported lower avidity and antibodies after (3rd) booster vaccination [16]. Conflicting results may be due to different methodologies or might be related to the later time point of analysis and waning antibody levels. However, data from a kinetic study in CVID patients showed relatively stable anti-spike IgG antibody levels 4 weeks and 20 weeks after 2nd COVID-19 vaccination and a positive trend for increased avidity after 3rd vaccination [9]. Immunological and genetic heterogeneity within the group of COVID-19 seroresponding CVID patients are likely to contribute to the different observations.
SARS-CoV-2-specific IgG antibodies in peripheral blood are not informative about the source or MBC functionality and may arise from short- or long-lived plasma cells or from MBC after differentiation into ASC.
While conventional ELISPOT assay provides a qualitative and quantitative readout and can be designed to detect specific antibody responses [23, 24], the use of ELISPOT following in vitro stimulation and differentiation of MBC into ASC enables a functional analysis of specific B cell memory. Using an in-house SARS-CoV-2 spike–specific MBC ELISPOT assay, all HC showed a detectable response; however, only 3 (30%) seroresponding CVID patients had minimally detectable SARS-CoV-2 spike–specific IgG from ASCs after in vitro simulation and differentiation. Ratio of specific ASC per CS MBC as well as per CS PB was as significantly lower in CVID patients, indicating that the majority of seroresponding CVID patients failed to develop a robust humoral memory response.
Data on B cell memory in CVID patients after COVID-19 vaccination are very limited. Using flow cytometry, SARS-CoV-2–specific atypical MBC (defined as CD19+CD24CD27CD38) with proposed low affinity were reported [17]. The present study provides additional functional data, showing an impaired specific recall memory response in seroresponding CVID patients. In the general population, SARS-CoV-2 mRNA-based vaccination induces both, a persistent germinal center (GC) B cell response and a robust but transient extra-follicular (EF) immune response resulting in antibodies of lower affinity from circulating PB [25, 26]. Lower avidity and impaired humoral memory formation argue for a predominantly EF and impaired GC response in our cohort of COVID-19-vaccinated CVID patients.
In addition to B cell differentiation and maturation, GC reaction involves multiple B cell extrinsic factors including specific (follicular) T cell interactions. Previous data suggested a correlation between reduced specific humoral immune response and impaired specific T cellular immunity in CVID patients [17, 27]. Our findings contrast this observation, with all CVID seroresponder and 5/6 non-seroresponder showing a robust polyfunctional CD4+ T cell immune response thus complementing previous reports of robust specific SARS-CoV-2 T cell responses in CVID patients with mild [28] and severe SARS-CoV-2 infections [29] as well as to COVID-19 vaccination [810] and other vaccines, such as influenza [30, 31]. A limitation of our study is the relatively long period of collecting samples in CVID patients and HC. While healthy control was analyzed earlier, all participating individuals were evaluated at least 4 weeks after 2nd vaccination. Multiple studies show a stable specific SARS-CoV-2 CD4 + cellular immune response between 2 weeks and 6 months after second COVID-19 vaccination in healthy individuals [9, 32, 33] and for CVID patients [8].
The relatively broad range in specific T cell immunity in CVID patients may at least partly attributable to the applied methodology of analyzing SARS-CoV-2-specific T cell immunity. This is exemplified in our cohort by the variability of T cell responses ranging from 31 to 44% no or low responders when using commercially available IGRA assay and reaching 94% patients with polyfunctional triple-positive activated T cells by flow cytometry. In addition to methodological aspects, the general clinical and immunological heterogeneity of CVID patients may help to reconcile different observations. Higher frequencies of activated T cells in non-seroresponding CVID patients to SEB as positive control challenge the hypothesis of a generally impaired T cell immunity leading to a lower specific T cell response in our cohort of CVID patients [13].
TFH cell and B cell interaction during GC reaction are a prerequisite for high-affinity antibody formation and levels of specific TFH were reported to correlate positively with vaccine-induced antibodies against conjugated pneumococcal, hepatitis B, and influenza [34, 35]. In SARS-CoV-2, mRNA vaccination was shown to induce a robust specific TFH response with stable persistence for at least 6 months after 2nd vaccination [36].
The role of TFH cells in CVID patients remains poorly understand. While a preserved TFH response was observed upon influenza vaccination [31], lower levels of specific TFH cells were reported in a cohort of COVID-19-vaccinated CVID patients [13]; however, analysis of TFH cell response was not differentiated into seroresponding and seronegative patients in this study. In our cohort, we could successfully identify a TFH response after stimulation with SARS-CoV-2 spike peptide pools with CVID seroresponder and HC expressing similar frequencies. However, in seronegative CVID patients, TFH cells did not increase significantly after stimulation with N-terminal spike and also TFH response to C-terminal SARS-CoV-2 spike peptide pool was significantly lower in seronegative than in seroresponding CVID patients. This observation suggests an important role of TFH cells during COVID-19 vaccine response. However, we did not observe a correlation between levels of specific TFH cells with antibody levels or avidity.
Higher levels of activated CXCR5+ peripheral TFH cells were reported previously for CVID patients [37] in particular in patients with non-infectious manifestations (autoimmunity and granulomatous disease), suggesting a functional significance of this association. However, TFH cells form a functionally and phenotypically heterogeneous group. Although high expression of CXCR5 is one of the defining hallmarks of TFH, CXCR5 is also expressed on 20–25% of peripheral blood human central memory CD4+ T cells [38]. Detection of this subgroup prior to SARS-CoV-2 spike stimulation may therefore represent an unspecific state of activation. Of note, in CVID patients, peripheral TFH cells with phenotypical markers of activation were recently shown to express an mRNA signature of exhaustion, apoptosis, and senescence [39]. These observations should prompt caution when interpreting phenotypical findings in the context of TFH functionality. To further understand the role of TFH cells would require a detailed analysis, including functional assays and an expanded marker profile, in larger cohorts. Given the heterogeneity of changes in GC reactions in CVID patients, a uniform pathomechanism of impaired COVID-19 vaccine response is unlikely [4044].
Limitations of our study include the slight heterogeneity and timing of administered COVID-19 vaccines and that interpretation is restricted to patients after receiving two COVID-19 vaccinations. While more than two vaccinations were shown to increase humoral immune response in CVID patients [18], it remains uncertain whether repeated vaccinations could also translate into B cell memory formation. Due to increased levels of SARS-CoV-2-IgG antibodies in commercially available immunoglobulins [45], a mere serological evaluation does no longer allow to distinguish between passive immunization and active antibody generation. Using SARS-CoV-2-specific MBC ELISPOT assays may enable researchers to circumvent this uncertainty. HC were significantly younger; however by providing data before and after vaccination, we were able to analyze comparatively intra-individual responses within each group.
The identification of possible predictive factors of a strong or impaired immune response to COVID-19 vaccination would improve risk stratification and support an individual prophylactic management for CVID patients. Previous vaccination studies in CVID patients described the potential impact of the type of vaccine and distribution of B cell subsets affecting vaccination responses [46]. Regarding COVID-19 vaccine response in CVID, a range of immunological and clinical factors have been described, including non-infectious complications and ongoing immunosuppressive therapy as well as elevated CD21low B cells, low B cells, low naïve T cells, and reduced IgA and IgM levels [7, 11, 47]. In our cohort, CVID seroresponder and non-seroresponder did not differ in any key immunological parameter; however the small number of patients limits the interpretation. Of note, and in line with recent observations [9], previous failure to mount a specific antibody response to pneumococcal conjugate vaccine could not predict COVID-19 vaccine response in our cohort of CVID patients. This discrepancy might be related to the antigen structure, but immunogenicity might also depend on the mode of immunization (mRNA vs conjugated vaccine) [48]. While humoral immune response to pneumococcal conjugated vaccine requires GC reaction, response to COVID-19 involves both GC and EF structures [49]. Immunological and genetic aspects that shape the humoral immune response to SARS-CoV-2 vaccination in CVID patients remain incompletely understood. However, the observation of seroresponding and non-seroresponding CVID patients may help to shed light onto the diverse pathomechanisms of CVID. Further studies in larger cohorts are required to evaluate possible underlying B cell differentiation defects in CVID patients.

Conclusions

Reduced avidity of SARS-CoV-2 IgG and significantly impaired recall memory formation after COVID-19 vaccination in seroresponding CVID patients stress the importance of a more differentiated analysis of humoral immune response in CVID patients. Our observations challenge the binary categorization into seroresponder and non-seroresponder and potentially impact on clinical decisions for the prophylactic management of COVID-19.

Acknowledgements

We thank all patients and healthy controls who voluntarily donated their blood for this study. We acknowledge Marie Luisa Schmidt, Patricia Tscheak, and Petra Mackeldanz for excellent technical assistance.

Declarations

Ethics Approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the Ethics Committee of Charité, Universitätsmedizin Berlin (EA2/092/20 from June 4th 2020) and with the 1964 Helsinki Declaration and its later amendments.
Informed consent was obtained from all individual participants included in the study.
The authors affirm that human research participants provided informed consent for publication of acquired data.

Conflict of Interest

VMC is named together with Charité—Universitätsmedizin Berlin and Euroimmun GmbH on a patent application (Methods and reagents for diagnosis of SARS-CoV-2 infection. Pub number 20210190797) filed recently regarding the diagnostic of SARS-CoV-2 by antibody testing. The other authors have no relevant financial or non-financial interests to disclose.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
2.
Zurück zum Zitat Milito C, Cinetto F, Palladino A, Garzi G, Punziano A, Lagnese G, et al. Mortality in severe antibody deficiencies patients during the first two years of the COVID-19 pandemic: vaccination and monoclonal antibodies efficacy. Biomedicines. 2022;10(5):1026.PubMedPubMedCentralCrossRef Milito C, Cinetto F, Palladino A, Garzi G, Punziano A, Lagnese G, et al. Mortality in severe antibody deficiencies patients during the first two years of the COVID-19 pandemic: vaccination and monoclonal antibodies efficacy. Biomedicines. 2022;10(5):1026.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Katzenstein TL, Rasmussen LD, Drabe CH, Larsen CS, Hansen AE, Stærkind M, et al. Outcome of SARS-CoV-2 infection among patients with common variable immunodeficiency and a matched control group: A Danish nationwide cohort study. Front Immunol. 2022;13: 994253.PubMedPubMedCentralCrossRef Katzenstein TL, Rasmussen LD, Drabe CH, Larsen CS, Hansen AE, Stærkind M, et al. Outcome of SARS-CoV-2 infection among patients with common variable immunodeficiency and a matched control group: A Danish nationwide cohort study. Front Immunol. 2022;13: 994253.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Milito C, Cinetto F, Garzi G, Palladino A, Puca M, Brambilla E, et al. Safety of mRNA COVID-19 vaccines in patients with inborn errors of immunity: an Italian multicentric study. J Clin Immunol. 2023;43(2):299–307.PubMedCrossRef Milito C, Cinetto F, Garzi G, Palladino A, Puca M, Brambilla E, et al. Safety of mRNA COVID-19 vaccines in patients with inborn errors of immunity: an Italian multicentric study. J Clin Immunol. 2023;43(2):299–307.PubMedCrossRef
5.
Zurück zum Zitat Delmonte OM, Castagnoli R, Notarangelo LD. COVID-19 and inborn errors of immunity. Physiology (Bethesda). 2022;37(6):0. Delmonte OM, Castagnoli R, Notarangelo LD. COVID-19 and inborn errors of immunity. Physiology (Bethesda). 2022;37(6):0.
6.
Zurück zum Zitat Hagin D, Freund T, Navon M, Halperin T, Adir D, Marom R, et al. Immunogenicity of Pfizer-BioNTech COVID-19 vaccine in patients with inborn errors of immunity. J Allergy Clin Immunol. 2021;148(3):739–49.PubMedPubMedCentralCrossRef Hagin D, Freund T, Navon M, Halperin T, Adir D, Marom R, et al. Immunogenicity of Pfizer-BioNTech COVID-19 vaccine in patients with inborn errors of immunity. J Allergy Clin Immunol. 2021;148(3):739–49.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat van Leeuwen LPM, GeurtsvanKessel CH, Ellerbroek PM, de Bree GJ, Potjewijd J, Rutgers A, et al. Immunogenicity of the mRNA-1273 COVID-19 vaccine in adult patients with inborn errors of immunity. J Allergy Clin Immunol. 2022;149(6):1949–57.PubMedPubMedCentralCrossRef van Leeuwen LPM, GeurtsvanKessel CH, Ellerbroek PM, de Bree GJ, Potjewijd J, Rutgers A, et al. Immunogenicity of the mRNA-1273 COVID-19 vaccine in adult patients with inborn errors of immunity. J Allergy Clin Immunol. 2022;149(6):1949–57.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Milota T, Smetanova J, Skotnicova A, Rataj M, Lastovicka J, Zelena H, et al. Clinical outcomes, immunogenicity, and safety of BNT162b2 vaccine in primary antibody deficiency. J Allergy Clin Immunol Pract. 2023;11(1):306-14.e2.PubMedCrossRef Milota T, Smetanova J, Skotnicova A, Rataj M, Lastovicka J, Zelena H, et al. Clinical outcomes, immunogenicity, and safety of BNT162b2 vaccine in primary antibody deficiency. J Allergy Clin Immunol Pract. 2023;11(1):306-14.e2.PubMedCrossRef
9.
Zurück zum Zitat Ainsua-Enrich E, Pedreño-Lopez N, Bracke C, Ávila-Nieto C, Rodríguez dela Concepción ML, Pradenas E, et al. Kinetics of immune responses elicited after three mRNA COVID-19 vaccine doses in predominantly antibody-deficient individuals. iScience. 2022;25(11):105455.PubMedPubMedCentralCrossRef Ainsua-Enrich E, Pedreño-Lopez N, Bracke C, Ávila-Nieto C, Rodríguez dela Concepción ML, Pradenas E, et al. Kinetics of immune responses elicited after three mRNA COVID-19 vaccine doses in predominantly antibody-deficient individuals. iScience. 2022;25(11):105455.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Goda V, Kriván G, Kulcsár A, Gönczi M, Tasnády S, Matula Z, et al. Specific antibody and the T-cell response elicited by BNT162b2 boosting after two ChAdOx1 nCoV-19 in common variable immunodeficiency. Front Immunol. 2022;13: 907125.PubMedPubMedCentralCrossRef Goda V, Kriván G, Kulcsár A, Gönczi M, Tasnády S, Matula Z, et al. Specific antibody and the T-cell response elicited by BNT162b2 boosting after two ChAdOx1 nCoV-19 in common variable immunodeficiency. Front Immunol. 2022;13: 907125.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Shin JJ, Par-Young J, Unlu S, McNamara A, Park HJ, Shin MS, et al. Defining clinical and immunological predictors of poor immune responses to COVID-19 mRNA vaccines in patients with primary antibody deficiency. J Clin Immunol. 2022;42(6):1137–50.PubMedPubMedCentralCrossRef Shin JJ, Par-Young J, Unlu S, McNamara A, Park HJ, Shin MS, et al. Defining clinical and immunological predictors of poor immune responses to COVID-19 mRNA vaccines in patients with primary antibody deficiency. J Clin Immunol. 2022;42(6):1137–50.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Shields AM, Faustini SE, Hill HJ, Al-Taei S, Tanner C, Ashford F, et al. SARS-CoV-2 vaccine responses in individuals with antibody deficiency: findings from the COV-AD study. J Clin Immunol. 2022;42(5):923–34.PubMedPubMedCentralCrossRef Shields AM, Faustini SE, Hill HJ, Al-Taei S, Tanner C, Ashford F, et al. SARS-CoV-2 vaccine responses in individuals with antibody deficiency: findings from the COV-AD study. J Clin Immunol. 2022;42(5):923–34.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Sauerwein KMT, Geier CB, Stemberger RF, Akyaman H, Illes P, Fischer MB, et al. Antigen-specific CD4(+) T-cell activation in primary antibody deficiency after BNT162b2 mRNA COVID-19 vaccination. Front Immunol. 2022;13: 827048.PubMedPubMedCentralCrossRef Sauerwein KMT, Geier CB, Stemberger RF, Akyaman H, Illes P, Fischer MB, et al. Antigen-specific CD4(+) T-cell activation in primary antibody deficiency after BNT162b2 mRNA COVID-19 vaccination. Front Immunol. 2022;13: 827048.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Arroyo-Sánchez D, Cabrera-Marante O, Laguna-Goya R, Almendro-Vázquez P, Carretero O, Gil-Etayo FJ, et al. Immunogenicity of anti-SARS-CoV-2 vaccines in common variable immunodeficiency. J Clin Immunol. 2022;42(2):240–52.PubMedCrossRef Arroyo-Sánchez D, Cabrera-Marante O, Laguna-Goya R, Almendro-Vázquez P, Carretero O, Gil-Etayo FJ, et al. Immunogenicity of anti-SARS-CoV-2 vaccines in common variable immunodeficiency. J Clin Immunol. 2022;42(2):240–52.PubMedCrossRef
15.
Zurück zum Zitat Göschl L, Mrak D, Grabmeier-Pfistershammer K, Stiasny K, Haslacher H, Schneider L, et al. Reactogenicity and immunogenicity of the second COVID-19 vaccination in patients with inborn errors of immunity or mannan-binding lectin deficiency. Front Immunol. 2022;13: 974987.PubMedPubMedCentralCrossRef Göschl L, Mrak D, Grabmeier-Pfistershammer K, Stiasny K, Haslacher H, Schneider L, et al. Reactogenicity and immunogenicity of the second COVID-19 vaccination in patients with inborn errors of immunity or mannan-binding lectin deficiency. Front Immunol. 2022;13: 974987.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Salinas AF, Mortari EP, Terreri S, Quintarelli C, Pulvirenti F, Di Cecca S, et al. SARS-CoV-2 vaccine induced atypical immune responses in antibody defects: everybody does their best. J Clin Immunol. 2021;41(8):1709–22.PubMedPubMedCentralCrossRef Salinas AF, Mortari EP, Terreri S, Quintarelli C, Pulvirenti F, Di Cecca S, et al. SARS-CoV-2 vaccine induced atypical immune responses in antibody defects: everybody does their best. J Clin Immunol. 2021;41(8):1709–22.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Nielsen BU, Drabe CH, Barnkob MB, Johansen IS, Hansen AKK, Nilsson AC, et al. Antibody response following the third and fourth SARS-CoV-2 vaccine dose in individuals with common variable immunodeficiency. Front Immunol. 2022;13: 934476.PubMedPubMedCentralCrossRef Nielsen BU, Drabe CH, Barnkob MB, Johansen IS, Hansen AKK, Nilsson AC, et al. Antibody response following the third and fourth SARS-CoV-2 vaccine dose in individuals with common variable immunodeficiency. Front Immunol. 2022;13: 934476.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Seidel MG, Kindle G, Gathmann B, Quinti I, Buckland M, van Montfrans J, et al. The European Society for Immunodeficiencies (ESID) registry working definitions for the clinical diagnosis of inborn errors of immunity. J Allergy Clin Immunol Pract. 2019;7(6):1763–70.PubMedCrossRef Seidel MG, Kindle G, Gathmann B, Quinti I, Buckland M, van Montfrans J, et al. The European Society for Immunodeficiencies (ESID) registry working definitions for the clinical diagnosis of inborn errors of immunity. J Allergy Clin Immunol Pract. 2019;7(6):1763–70.PubMedCrossRef
20.
Zurück zum Zitat Schwarz T, Otto C, Jones TC, Pache F, Schindler P, Niederschweiberer M, et al. Preserved T cell responses to SARS-CoV-2 in anti-CD20 treated multiple sclerosis. Mult Scler. 2022;28(7):1041–50.PubMedPubMedCentralCrossRef Schwarz T, Otto C, Jones TC, Pache F, Schindler P, Niederschweiberer M, et al. Preserved T cell responses to SARS-CoV-2 in anti-CD20 treated multiple sclerosis. Mult Scler. 2022;28(7):1041–50.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Crotty S, Aubert RD, Glidewell J, Ahmed R. Tracking human antigen-specific memory B cells: a sensitive and generalized ELISPOT system. J Immunol Methods. 2004;286(1–2):111–22.PubMedCrossRef Crotty S, Aubert RD, Glidewell J, Ahmed R. Tracking human antigen-specific memory B cells: a sensitive and generalized ELISPOT system. J Immunol Methods. 2004;286(1–2):111–22.PubMedCrossRef
22.
Zurück zum Zitat Wehr C, Kivioja T, Schmitt C, Ferry B, Witte T, Eren E, et al. The EUROclass trial: defining subgroups in common variable immunodeficiency. Blood. 2008;111(1):77–85.PubMedCrossRef Wehr C, Kivioja T, Schmitt C, Ferry B, Witte T, Eren E, et al. The EUROclass trial: defining subgroups in common variable immunodeficiency. Blood. 2008;111(1):77–85.PubMedCrossRef
23.
Zurück zum Zitat Tarkowski A, Czerkinsky C, Nilsson LA, Nygren H, Ouchterlony O. Solid-phase enzyme-linked immunospot (ELISPOT) assay for enumeration of IgG rheumatoid factor-secreting cells. J Immunol Methods. 1984;72(2):451–9.PubMedCrossRef Tarkowski A, Czerkinsky C, Nilsson LA, Nygren H, Ouchterlony O. Solid-phase enzyme-linked immunospot (ELISPOT) assay for enumeration of IgG rheumatoid factor-secreting cells. J Immunol Methods. 1984;72(2):451–9.PubMedCrossRef
24.
Zurück zum Zitat Chovancova Z, Vlkova M, Litzman J, Lokaj J, Thon V. Antibody forming cells and plasmablasts in peripheral blood in CVID patients after vaccination. Vaccine. 2011;29(24):4142–50.PubMedCrossRef Chovancova Z, Vlkova M, Litzman J, Lokaj J, Thon V. Antibody forming cells and plasmablasts in peripheral blood in CVID patients after vaccination. Vaccine. 2011;29(24):4142–50.PubMedCrossRef
25.
Zurück zum Zitat Turner JS, O’Halloran JA, Kalaidina E, Kim W, Schmitz AJ, Zhou JQ, et al. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses. Nature. 2021;596(7870):109–13.PubMedPubMedCentralCrossRef Turner JS, O’Halloran JA, Kalaidina E, Kim W, Schmitz AJ, Zhou JQ, et al. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses. Nature. 2021;596(7870):109–13.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Kim W, Zhou JQ, Horvath SC, Schmitz AJ, Sturtz AJ, Lei T, et al. Germinal centre-driven maturation of B cell response to mRNA vaccination. Nature. 2022;604(7904):141–5.PubMedPubMedCentralCrossRef Kim W, Zhou JQ, Horvath SC, Schmitz AJ, Sturtz AJ, Lei T, et al. Germinal centre-driven maturation of B cell response to mRNA vaccination. Nature. 2022;604(7904):141–5.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Pulvirenti F, Di Cecca S, Sinibaldi M, Piano Mortari E, Terreri S, Albano C, et al. T-cell defects associated to lack of spike-specific antibodies after BNT162b2 full immunization followed by a booster dose in patients with common variable immune deficiencies. Cells. 2022;11(12):1918.PubMedPubMedCentralCrossRef Pulvirenti F, Di Cecca S, Sinibaldi M, Piano Mortari E, Terreri S, Albano C, et al. T-cell defects associated to lack of spike-specific antibodies after BNT162b2 full immunization followed by a booster dose in patients with common variable immune deficiencies. Cells. 2022;11(12):1918.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Kinoshita H, Durkee-Shock J, Jensen-Wachspress M, Kankate VV, Lang H, Lazarski CA, et al. Robust antibody and T cell responses to SARS-CoV-2 in patients with antibody deficiency. J Clin Immunol. 2021;41(6):1146–53.PubMedPubMedCentralCrossRef Kinoshita H, Durkee-Shock J, Jensen-Wachspress M, Kankate VV, Lang H, Lazarski CA, et al. Robust antibody and T cell responses to SARS-CoV-2 in patients with antibody deficiency. J Clin Immunol. 2021;41(6):1146–53.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Steiner S, Schwarz T, Corman VM, Gebert L, Kleinschmidt MC, Wald A, et al. SARS-CoV-2 T cell response in severe and fatal COVID-19 in primary antibody deficiency patients without specific humoral immunity. Front Immunol. 2022;13:840126. Steiner S, Schwarz T, Corman VM, Gebert L, Kleinschmidt MC, Wald A, et al. SARS-CoV-2 T cell response in severe and fatal COVID-19 in primary antibody deficiency patients without specific humoral immunity. Front Immunol. 2022;13:840126.
30.
Zurück zum Zitat Hanitsch LG, Lobel M, Mieves JF, Bauer S, Babel N, Schweiger B, et al. Cellular and humoral influenza-specific immune response upon vaccination in patients with common variable immunodeficiency and unclassified antibody deficiency. Vaccine. 2016;34(21):2417–23.PubMedCrossRef Hanitsch LG, Lobel M, Mieves JF, Bauer S, Babel N, Schweiger B, et al. Cellular and humoral influenza-specific immune response upon vaccination in patients with common variable immunodeficiency and unclassified antibody deficiency. Vaccine. 2016;34(21):2417–23.PubMedCrossRef
31.
Zurück zum Zitat Friedmann D, Goldacker S, Peter HH, Warnatz K. Preserved cellular immunity upon influenza vaccination in most patients with common variable immunodeficiency. J Allergy Clin Immunol Pract. 2020;8(7):2332-40.e5.PubMedCrossRef Friedmann D, Goldacker S, Peter HH, Warnatz K. Preserved cellular immunity upon influenza vaccination in most patients with common variable immunodeficiency. J Allergy Clin Immunol Pract. 2020;8(7):2332-40.e5.PubMedCrossRef
32.
Zurück zum Zitat Painter MM, Mathew D, Goel RR, Apostolidis SA, Pattekar A, Kuthuru O, et al. Rapid induction of antigen-specific CD4(+) T cells is associated with coordinated humoral and cellular immunity to SARS-CoV-2 mRNA vaccination. Immunity. 2021;54(9):2133-42.e3.PubMedPubMedCentralCrossRef Painter MM, Mathew D, Goel RR, Apostolidis SA, Pattekar A, Kuthuru O, et al. Rapid induction of antigen-specific CD4(+) T cells is associated with coordinated humoral and cellular immunity to SARS-CoV-2 mRNA vaccination. Immunity. 2021;54(9):2133-42.e3.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Collier AY, Yu J, McMahan K, Liu J, Chandrashekar A, Maron JS, et al. Differential kinetics of immune responses elicited by Covid-19 vaccines. N Engl J Med. 2021;385(21):2010–2.PubMedCrossRef Collier AY, Yu J, McMahan K, Liu J, Chandrashekar A, Maron JS, et al. Differential kinetics of immune responses elicited by Covid-19 vaccines. N Engl J Med. 2021;385(21):2010–2.PubMedCrossRef
34.
Zurück zum Zitat Xing M, Feng Y, Yao J, Lv H, Chen Y, He H, et al. Induction of peripheral blood T follicular helper cells expressing ICOS correlates with antibody response to hepatitis B vaccination. J Med Virol. 2020;92(1):62–70.PubMedCrossRef Xing M, Feng Y, Yao J, Lv H, Chen Y, He H, et al. Induction of peripheral blood T follicular helper cells expressing ICOS correlates with antibody response to hepatitis B vaccination. J Med Virol. 2020;92(1):62–70.PubMedCrossRef
35.
Zurück zum Zitat Sterrett S, Peng BJ, Burton RL, LaFon DC, Westfall AO, Singh S, et al. Peripheral CD4 T follicular cells induced by a conjugated pneumococcal vaccine correlate with enhanced opsonophagocytic antibody responses in younger individuals. Vaccine. 2020;38(7):1778–86.PubMedPubMedCentralCrossRef Sterrett S, Peng BJ, Burton RL, LaFon DC, Westfall AO, Singh S, et al. Peripheral CD4 T follicular cells induced by a conjugated pneumococcal vaccine correlate with enhanced opsonophagocytic antibody responses in younger individuals. Vaccine. 2020;38(7):1778–86.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Mudd PA, Minervina AA, Pogorelyy MV, Turner JS, Kim W, Kalaidina E, et al. SARS-CoV-2 mRNA vaccination elicits a robust and persistent T follicular helper cell response in humans. Cell. 2022;185(4):603-13.e15.PubMedCrossRef Mudd PA, Minervina AA, Pogorelyy MV, Turner JS, Kim W, Kalaidina E, et al. SARS-CoV-2 mRNA vaccination elicits a robust and persistent T follicular helper cell response in humans. Cell. 2022;185(4):603-13.e15.PubMedCrossRef
37.
Zurück zum Zitat Coraglia A, Galassi N, Fernández Romero DS, Juri MC, Felippo M, Malbrán A, et al. Common variable immunodeficiency and circulating TFH. J Immunol Res. 2016;2016:4951587.PubMedPubMedCentralCrossRef Coraglia A, Galassi N, Fernández Romero DS, Juri MC, Felippo M, Malbrán A, et al. Common variable immunodeficiency and circulating TFH. J Immunol Res. 2016;2016:4951587.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Chevalier N, Jarrossay D, Ho E, Avery DT, Ma CS, Yu D, et al. CXCR5 expressing human central memory CD4 T cells and their relevance for humoral immune responses. J Immunol. 2011;186(10):5556–68.PubMedCrossRef Chevalier N, Jarrossay D, Ho E, Avery DT, Ma CS, Yu D, et al. CXCR5 expressing human central memory CD4 T cells and their relevance for humoral immune responses. J Immunol. 2011;186(10):5556–68.PubMedCrossRef
39.
Zurück zum Zitat Milardi G, Di Lorenzo B, Gerosa J, Barzaghi F, Di Matteo G, Omrani M, et al. Follicular helper T cell signature of replicative exhaustion, apoptosis, and senescence in common variable immunodeficiency. Eur J Immunol. 2022;52(7):1171–89.PubMedPubMedCentralCrossRef Milardi G, Di Lorenzo B, Gerosa J, Barzaghi F, Di Matteo G, Omrani M, et al. Follicular helper T cell signature of replicative exhaustion, apoptosis, and senescence in common variable immunodeficiency. Eur J Immunol. 2022;52(7):1171–89.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Unger S, Seidl M, Schmitt-Graeff A, Böhm J, Schrenk K, Wehr C, et al. Ill-defined germinal centers and severely reduced plasma cells are histological hallmarks of lymphadenopathy in patients with common variable immunodeficiency. J Clin Immunol. 2014;34(6):615–26.PubMedCrossRef Unger S, Seidl M, Schmitt-Graeff A, Böhm J, Schrenk K, Wehr C, et al. Ill-defined germinal centers and severely reduced plasma cells are histological hallmarks of lymphadenopathy in patients with common variable immunodeficiency. J Clin Immunol. 2014;34(6):615–26.PubMedCrossRef
41.
Zurück zum Zitat Romberg N, Le Coz C, Glauzy S, Schickel JN, Trofa M, Nolan BE, et al. Patients with common variable immunodeficiency with autoimmune cytopenias exhibit hyperplastic yet inefficient germinal center responses. J Allergy Clin Immunol. 2019;143(1):258–65.PubMedCrossRef Romberg N, Le Coz C, Glauzy S, Schickel JN, Trofa M, Nolan BE, et al. Patients with common variable immunodeficiency with autoimmune cytopenias exhibit hyperplastic yet inefficient germinal center responses. J Allergy Clin Immunol. 2019;143(1):258–65.PubMedCrossRef
42.
Zurück zum Zitat Del Pino Molina L, Torres Canizales JM, Pernía O, Rodríguez Pena R, Ibanez de Caceres I, López Granados E. Defective Bcl-2 expression in memory B cells from common variable immunodeficiency patients. Clin Exp Immunol. 2021;203(3):341–50.PubMedCrossRef Del Pino Molina L, Torres Canizales JM, Pernía O, Rodríguez Pena R, Ibanez de Caceres I, López Granados E. Defective Bcl-2 expression in memory B cells from common variable immunodeficiency patients. Clin Exp Immunol. 2021;203(3):341–50.PubMedCrossRef
43.
Zurück zum Zitat van Schouwenburg P, Unger S, Payne KJ, Kaiser FMP, Pico-Knijnenburg I, Pfeiffer J, et al. Deciphering imprints of impaired memory B-cell maturation in germinal centers of three patients with common variable immunodeficiency. Front Immunol. 2022;13: 959002.PubMedPubMedCentralCrossRef van Schouwenburg P, Unger S, Payne KJ, Kaiser FMP, Pico-Knijnenburg I, Pfeiffer J, et al. Deciphering imprints of impaired memory B-cell maturation in germinal centers of three patients with common variable immunodeficiency. Front Immunol. 2022;13: 959002.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Driessen GJ, van Zelm MC, van Hagen PM, Hartwig NG, Trip M, Warris A, et al. B-cell replication history and somatic hypermutation status identify distinct pathophysiologic backgrounds in common variable immunodeficiency. Blood. 2011;118(26):6814–23.PubMedCrossRef Driessen GJ, van Zelm MC, van Hagen PM, Hartwig NG, Trip M, Warris A, et al. B-cell replication history and somatic hypermutation status identify distinct pathophysiologic backgrounds in common variable immunodeficiency. Blood. 2011;118(26):6814–23.PubMedCrossRef
45.
Zurück zum Zitat Hirsiger JR, Weigang S, Walz AC, Fuchs J, Daly ML, Eggimann S, et al. Passive immunization against COVID-19 by anti-SARS-CoV-2 spike IgG in commercially available immunoglobulin preparations in severe antibody deficiency. J Allergy Clin Immunol Pract. 2022;10(9):2452-5.e3.PubMedPubMedCentralCrossRef Hirsiger JR, Weigang S, Walz AC, Fuchs J, Daly ML, Eggimann S, et al. Passive immunization against COVID-19 by anti-SARS-CoV-2 spike IgG in commercially available immunoglobulin preparations in severe antibody deficiency. J Allergy Clin Immunol Pract. 2022;10(9):2452-5.e3.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Goldacker S, Draeger R, Warnatz K, Huzly D, Salzer U, Thiel J, et al. Active vaccination in patients with common variable immunodeficiency (CVID). Clin Immunol. 2007;124(3):294–303.PubMedCrossRef Goldacker S, Draeger R, Warnatz K, Huzly D, Salzer U, Thiel J, et al. Active vaccination in patients with common variable immunodeficiency (CVID). Clin Immunol. 2007;124(3):294–303.PubMedCrossRef
47.
Zurück zum Zitat Bergman P, Wullimann D, Gao Y, Wahren Borgström E, Norlin AC, Lind Enoksson S, et al. Elevated CD21(low) B cell frequency is a marker of poor immunity to Pfizer-BioNTech BNT162b2 mRNA vaccine against SARS-CoV-2 in patients with common variable immunodeficiency. J Clin Immunol. 2022;42(4):716–27.PubMedPubMedCentralCrossRef Bergman P, Wullimann D, Gao Y, Wahren Borgström E, Norlin AC, Lind Enoksson S, et al. Elevated CD21(low) B cell frequency is a marker of poor immunity to Pfizer-BioNTech BNT162b2 mRNA vaccine against SARS-CoV-2 in patients with common variable immunodeficiency. J Clin Immunol. 2022;42(4):716–27.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Elsner RA, Shlomchik MJ. Germinal center and extrafollicular B cell responses in vaccination, immunity, and autoimmunity. Immunity. 2020;53(6):1136–50.PubMedPubMedCentralCrossRef Elsner RA, Shlomchik MJ. Germinal center and extrafollicular B cell responses in vaccination, immunity, and autoimmunity. Immunity. 2020;53(6):1136–50.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Woodruff MC, Ramonell RP, Nguyen DC, Cashman KS, Saini AS, Haddad NS, et al. Extrafollicular B cell responses correlate with neutralizing antibodies and morbidity in COVID-19. Nat Immunol. 2020;21(12):1506–16.PubMedPubMedCentralCrossRef Woodruff MC, Ramonell RP, Nguyen DC, Cashman KS, Saini AS, Haddad NS, et al. Extrafollicular B cell responses correlate with neutralizing antibodies and morbidity in COVID-19. Nat Immunol. 2020;21(12):1506–16.PubMedPubMedCentralCrossRef
Metadaten
Titel
Impaired B Cell Recall Memory and Reduced Antibody Avidity but Robust T Cell Response in CVID Patients After COVID-19 Vaccination
verfasst von
Sophie Steiner
Tatjana Schwarz
Victor M. Corman
Lara M. Jeworowski
Sandra Bauer
Christian Drosten
Carmen Scheibenbogen
Leif G. Hanitsch
Publikationsdatum
17.03.2023
Verlag
Springer US
Erschienen in
Journal of Clinical Immunology / Ausgabe 5/2023
Print ISSN: 0271-9142
Elektronische ISSN: 1573-2592
DOI
https://doi.org/10.1007/s10875-023-01468-w

Weitere Artikel der Ausgabe 5/2023

Journal of Clinical Immunology 5/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Triglyzeridsenker schützt nicht nur Hochrisikopatienten

10.05.2024 Hypercholesterinämie Nachrichten

Patienten mit Arteriosklerose-bedingten kardiovaskulären Erkrankungen, die trotz Statineinnahme zu hohe Triglyzeridspiegel haben, profitieren von einer Behandlung mit Icosapent-Ethyl, und zwar unabhängig vom individuellen Risikoprofil.

Gibt es eine Wende bei den bioresorbierbaren Gefäßstützen?

In den USA ist erstmals eine bioresorbierbare Gefäßstütze – auch Scaffold genannt – zur Rekanalisation infrapoplitealer Arterien bei schwerer PAVK zugelassen worden. Das markiert einen Wendepunkt in der Geschichte dieser speziellen Gefäßstützen.

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

Wie managen Sie die schmerzhafte diabetische Polyneuropathie?

10.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Mit Capsaicin-Pflastern steht eine neue innovative Therapie bei schmerzhafter diabetischer Polyneuropathie zur Verfügung. Bei therapierefraktären Schmerzen stellt die Hochfrequenz-Rückenmarkstimulation eine adäquate Option dar.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.