Introduction
Patients with kidney failure requiring hemodialysis have a high mortality rate after infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (1–3 ). Additionally, in-center hemodialysis is associated with a higher risk of contact with health professionals or other high-risk patients, which may explain the elevated incidence of SARS-CoV-2 infection observed in patients on dialysis (4 ,5 ). Consequently, the dialysis population appears as a paradigm of a high-risk population in the SARS-CoV-2 pandemic. The rapid vaccine rollout had a major role in controlling infection spread (6 ), but patients on dialysis have been largely excluded from vaccine trials for safety reasons. To date, characterization of SARS-CoV-2 vaccine response in patients on dialysis is mainly on the basis of serologic response, showing an 80%–95% rate of positive antispike IgG serology after mRNA vaccination (7 ), although antibody levels appeared reduced when compared with the nondialysis population (7–10 ). The analyses of vaccine efficacy in patients on dialysis suggest a clinical efficacy against coronavirus 2019 (COVID-19) (10 ,11 ), but this efficacy could be lower than in the general population because severe forms are not rare even after the complete vaccine scheme (two doses) (12 ). In parallel to the rapid serum IgG response, vaccination induces the progressive generation of memory B cells , which continuously acquire somatic mutations in their variable region genes to improve affinity and constitute another layer of immune protection. After antigenic re-exposure, memory B cells subsequently drive the recall response by differentiating into new antibody-secreting cells, providing high-affinity protective antibodies. In healthy individuals, although the antibody level wanes over time, memory B cells continued to increase in frequency and to mature between 3 and 6 months postvaccination (13 ,14 ) (R.R. Goel et al ., unpublished data), and thus, they constitute a key secondary layer of protection. Conversely, defective early humoral responses against SARS-CoV-2 have been reported in patients on dialysis and kidney transplant recipients (15 ).
The frailty of the dialysis population and concerns regarding long-term vaccine response (16 ) and clinical efficacy (12 ) have led to the recommendation of a third dose of vaccine since April 2021 for patients on dialysis in France. Several studies have shown an increase of antispike IgG response in virus-naive patients on dialysis after a third dose of mRNA vaccine (12 ,17–19 ), but the dynamics and persistence of the SARS-CoV-2–specific memory B cell pool after vaccination have not yet been characterized in the dialysis population. Similarly, the effect of a third vaccine dose in SARS-CoV-2–recovered individuals remains unclear, although patients who recovered from COVID-19 may now represent up to 20%–30% of the dialysis population in some areas (20 ). Detailed analyses of the vaccine response in this subgroup could have major importance in understanding the immunologic response after repeated antigen exposure.
Here, we characterized serologic and memory B cell response against the receptor-binding domain (RBD) region of the SARS-CoV-2 spike protein after mRNA vaccination in previously SARS-CoV-2–infected (SARS-CoV-2–recovered) and virus-naive patients on hemodialysis.
Materials and Methods
Patients on Dialysis
Fifty-nine patients on dialysis who received the BNT162b2 mRNA vaccine from three dialysis centers in the Ile-de-France region (Centre Hospitalier Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris, Creteil; Hôpital Privé Nord Parisien, Sarcelles; and Clinique de Villeneuve Saint Georges–Ramsay Sante, Villeneuve Saint Georges) were included in this study. Among them, 33 were naive to SARS-CoV-2 (virus naive), and 26 recovered from SARS-CoV-2 (SARS-CoV-2 recovered). Six SARS-CoV-2–recovered patients on dialysis had severe COVID-19 (hospitalized COVID-19 requiring oxygen), and 22 had mild COVID-19 (ambulatory disease). Median (minimum to maximum) time from COVID-19 diagnosis to first vaccine injection was 8 (2–11) months.
All patients on dialysis received at least two doses of the BNT162b2 mRNA vaccine at a mean time of 27.6 (±1.7) days after the first dose. Anti-RBD IgG titers and RBD-specific memory B cells were assessed 1–2 months after the second dose (anti-RBD IgG titers: mean 36 [SD±22] days after the second dose and RBD-specific memory B cells : mean 66 [SD±8] days after the second dose).
Among virus-naive patients on dialysis, 26 patients received a third dose at a median 145 (interquartile range [IQR], 70–153) days after the second dose and were sampled at a median of 50 (IQR, 45.5–50) days after the third dose.
Among SARS-CoV-2–recovered patients on dialysis, 13 received a third dose at a median of 145 (IQR, 70–154) days after the second dose and were sampled at a median of 49 (IQR, 42–50) days after the third injection. Five patients with only two vaccine injections were also sampled 6 months after the second dose (mean time 200 [SD±4] days from the second dose). Only one patient developed SARS-CoV-2 infection during follow-up. Vaccine policies were left to the discretion of the caring physician of the different centers, but no difference was observed in patients’ characteristics according to the number of vaccine doses.
The COVAC-HD study was approved by the ethical committee ELSAN institutional review board (2021-04-ATTIAS-01) and was performed in accordance with French law. Written informed consent was obtained from all participants.
Vaccinated Controls
Forty-nine individuals with no kidney disease (vaccinated controls) were included from the original MEMOCOV2 cohort (institutional review board 2018-A01610-55), including 23 healthy health care workers naive to SARS-CoV-2 (no history of COVID-19 and negative IgG antinucleocapsid and/or spike) and 26 patients recovered from mild or severe COVID-19 (SARS-CoV-2 recovered).
All vaccinated controls received the BNT162b2 mRNA vaccine. Virus-naive vaccinated controls received two doses at a mean of 27.7 days (SD±1.8 days) apart as part of the French vaccination campaign. SARS-CoV-2–recovered vaccinated controls received only one dose in line with French guidelines, except for three patients who received two doses. First injection was performed a mean of 308 days (SD±45 days) after the infection. Both cohorts were sampled 1–2 months after the first dose (SARS-CoV-2 recovered) or the second dose (SARS-CoV-2 recovered; mean ± SD: 64.05±13.3 and 63.3±9.0 days for naive, respectively).
Vaccinated control individuals were recruited at Henri Mondor University Hospital (Assistance Publique-Hôpitaux de Paris) between March and April 2021. The COVID-19: SARS-CoV-2 Specific Memory B and T-CD4+ Cells study (NCT04402892) was approved by the ethical committee of Ile-de-France VI (no. 40–20 HPS) and was performed in accordance with French law. Written informed consent was obtained from all participants.
Patients’ Characteristics
SARS-CoV-2 infection was defined as confirmed RT-PCR on nasal swab or clinical presentation associated with typical aspect on computed tomography scan and/or serologic evidence. SARS-CoV-2–naive individuals were defined as no history of COVID-19 and negative IgG antinucleocapsid (and/or spike). Immunocompromised patients were defined as patients with former kidney transplantation, current immunosuppressive treatment/chemotherapy or immunosuppressive treatment/chemotherapy discontinued for <6 months, or HIV infection (with CD4+ T cells below 500/mm3 ).
Statistical Analyses
Ordinary one-way ANOVA and Kruskal–Wallis tests were used to compare continuous variables as appropriate (indicated in Figures 1 and 2 ). Wilcoxon matched-pairs signed rank tests for longitudinal analysis were used. A P value =0.05 was considered statistically significant. Statistical analyses involved use of GraphPad Prism 9.0 (La Jolla, CA).
Figure 1.: Humoral and memory B cell response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor-binding domain (RBD) in virus-naive and SARS-CoV-2–recovered patients on dialysis after mRNA vaccination. (A) Antibody response. Median ± interquartile range (IQR) anti−SARS-CoV-2 RBD serum IgG titers measured by ELISA in virus-naive (left panel; white dots) and SARS-CoV-2–recovered individuals (right panel; mild coronavirus disease 2019 [COVID-19]: orange dots; severe COVID-19: red dots). White bars represent virus-naive (n =23) or SARS-CoV-2–recovered (n =26) healthy controls 1–2 months after two doses or one dose of mRNA vaccine, respectively (#). Light blue bars represent virus-naive (n =20) and SARS-CoV-2–recovered (n =20) patients on dialysis 1–2 months after a second dose of mRNA vaccine. Dark blue bars represent virus-naive (n =25) and SARS-CoV-2–recovered (n =13) patients on dialysis 1–2 months (median of 50 days) after a third dose of mRNA vaccine. (B) Antibody response. Longitudinal evolution of anti–SARS-CoV-2 RBD IgG titers after a third dose of mRNA vaccine in virus-naive (n =13) and SARS-CoV-2–recovered (n =7) patients on dialysis (median time from second dose to third dose: 153.5 [IQR, 145–155] days) and 6 months after a second dose in SARS-CoV-2–recovered (n =6) patients on dialysis. (C) Memory B cell response. Median ± IQR frequencies of SARS-CoV-2 RBD-specific cells in live CD19+ IgD− CD27+ CD38int/− memory B cells by flow cytometry in virus-naive and SARS-CoV-2–recovered individuals. (D) Memory B cell response. Frequencies of SARS-CoV-2 RBD-specific memory B cells after a third dose of mRNA vaccine in virus-naive (n =14) and SARS-CoV-2–recovered (n =7) patients on dialysis (median time from second dose to third dose: 153.5 [IQR, 145–155] days) and 6 months after a second dose in SARS-CoV-2–recovered (n =5) patients on dialysis. Kruskal–Wallis test with multiple comparisons between groups for (A) and (C) and Wilcoxon matched-pairs signed rank test for (B) and (D). Bars indicate median ± IQR. Values equal to zero were converted to 0.01 for logarithmic scale. **P <0.01; ***P <0.001; ****P <0.0001. 6M, 6 months; ns, nonsignificant; S, spike; A.U, arbitrary unit.
Figure 2.: Characterization of the anti-RBD B cell response. (A) Median ± IQR of frequencies of SARS-CoV-2 RBD-specific cells in live CD19+ IgD− CD27− CD38int/− DN B cells by flow cytometry in virus-naive (left panel; white dots) and SARS-CoV-2–recovered individuals (right panel; mild COVID-19: orange dots; severe COVID-19: red dots). White bars represent virus-naive (n =23) or SARS-CoV-2–recovered (n =23) healthy controls 1–2 months after two doses or one dose of mRNA vaccine, respectively (#). Light blue bars represent virus-naive (n =19) and SARS-CoV-2–recovered (n =17) patients on dialysis 1–2 months after a second dose of mRNA vaccine. Dark blue bars represent virus-naive (n =23) and SARS-CoV-2–recovered (n =13) patients on dialysis 1–2 months (median 50 days) after a third dose of mRNA vaccine. (B) Median ± IQR of frequencies of SARS-CoV-2 RBD-specific cells displaying a DN2 (CD19+ CD27− IgD− CD11c+ CD21− ) or a DN1 (CD19+ CD27− IgD− CD11c− CD21+ ) phenotype in virus-naive (n =9) and SARS-CoV-2–recovered (=18) healthy controls 1–2 months after two doses or one dose of mRNA vaccine, respectively; in virus-naive (n =9) and SARS-CoV-2–recovered (n =15) patients on dialysis 1–2 months after a second dose of mRNA vaccine; and in virus-naive (n =17) and SARS-CoV-2–recovered (n =9) patients on dialysis 1–2 months (median of 50 days) after a third dose of mRNA vaccine. We performed repeated measures mixed effects model analysis with multiple comparisons between time points for dialysis groups and Kruskal–Wallis with multiple comparisons between donor groups inside each time point and between vaccinated controls groups. Bars indicate median ± IQR. Values equal to zero were converted to 0.01 for logarithmic scale. *P <0.05; **P <0.01; ***P <0.001; ****P <0.0001. DN, double negative; S, spike; AU, arbitrary unit.
Detailed methods are provided in Supplemental Material .
Results
In total, 108 patients were followed in this study, including 59 patients on dialysis from three centers of the Paris area (Table 1 ). All patients received two doses (1 month apart) of the BNT162b2 vaccine (Pfizer/BioNTech) and then a third injection between 2 and 6 months after the first dose. We previously characterized the longitudinal evolution and maturation of SARS-CoV-2–responding B cells after mRNA vaccine in healthy individuals (21 ). Forty-nine individuals from this original cohort (virus naive or SARS-CoV-2 recovered) were included as controls and had all been sampled 1–2 months after the boost injection (second dose in virus-naive individuals and first dose in SARS-CoV-2–recovered individuals). Details of the study design and methodology are provided in Supplemental Material (Supplemental Figure 1 ).
Table 1. -
Clinical characteristics of vaccinated controls and patients on dialysis according to their severe acute respiratory syndrome coronavirus 2 infection status
Variable
Vaccinated Controls
Patients on Dialysis
Naive, n =23
Recovered, n =26
Naive, n =33
Recovered, n =26
Men, n (%)
11 (48)
9 (35)
23 (70)
17 (65)
Age, yr, median [IQR]
45 [33–54]
45 [36–59]
71 [67–77]
70 [65–73]
Comorbidities
Diabetes mellitus, n (%)
—
2 (8)
16 (48.5)
11 (42)
Hypertension, n (%)
—
3 (12)
32 (97)
25 (96)
Immunodepression, n (%)
—
0 (0)
5 (15)
2 (8)
Former kidney transplantation
1 (3)
1 (4)
Immunosuppressive therapy
2 (6)
1 (4)
Cancer with chemotherapy
1 (3)
HIV
1 (3)
Dialysis vintage, yr, median [IQR]
5.2 [1.9–6.4]
3.95 [2.7–5]
COVID-19 characteristic, n (%)
Mild presentation
—
20 (77)
—
19 (73)
Severe presentation
—
6 (23)
—
7 (27)
mRNA vaccine details
Sample available after 2 doses
21 (64)
20 (77)
Sample available after 3 doses
26 (79)
13 (50)
Sample available after 2 doses + 6 mo postboost
6 (23)
IQR, interquartile range; COVID-19, coronavirus disease 2019; —, no patients (0) had diabetes, hypertension, or immunodepression in naive vaccinated controls.
We first measured the pre- and postboost evolution of IgG serum titers against the RBD in virus-naive and SARS-CoV-2–recovered patients on dialysis (Supplemental Tables 2–5 ). We focused on the RBD region of the spike because anti-RBD antibodies contribute to the majority of neutralizing antibodies against SARS-CoV-2. After the second dose of mRNA vaccine, anti-RBD IgG response was quantitatively lower in virus-naive patients on dialysis compared with virus-naive controls (Figure 1A ) as previously reported (although the difference did not reach statistical significance in this cohort) (8 ,9 ). Strikingly, a third dose elicited a strong recall response, with anti-RBD IgG titers increasing, on average, by three-fold compared with titers postsecond dose (P =0.008) (Figure 1, A and B ). These levels were even higher than those of virus-naive nondialysis controls after two doses (P <0.001). In contrast, SARS-CoV-2–recovered patients on dialysis had reached elevated anti-RBD IgG serum titers after their second vaccine dose. These titers were higher than those of SARS-CoV-2–recovered vaccinated controls (who received only one dose according to French vaccine policy; P =0.005) (Figure 1A ). Moreover, in SARS-CoV-2–recovered patients on dialysis, the third dose did not lead to any significant increase in serum anti-RBD IgG titers (Figure 1, A and B ). We conclude that the third vaccine dose, when administered within 6 months, induced a strong recall response in virus-naive patients on dialysis but had no significant effect on anti-RBD IgG titers in SARS-CoV-2–recovered patients on dialysis.
To further explore these two distinct patterns of response, we next studied RBD-specific CD27+ IgD− memory B cells by flow cytometry as previously described (Supplemental Figure 2A , Supplemental Tables 1–5 ) (21 ).
We first analyzed the RBD-specific memory B cell frequencies after the second dose of mRNA vaccine in virus-naive individuals. Interestingly, we detected RBD-specific memory B cells at similar proportions in patients on dialysis and vaccinated controls, suggesting that the settlement of the memory B cell pool was not impaired in the virus-naive dialysis population. After a third dose of mRNA vaccine, we further observed a strong RBD-specific memory B cell expansion in virus-naive patients on dialysis with a 5.5-fold increase as compared with their frequencies after the second injection (P <0.001) (Figure 1, C and D ). We next studied SARS-CoV-2–recovered patients on dialysis. These patients already displayed robustly expanded RBD-specific memory B cells after two doses of mRNA vaccine, reaching frequencies similar to SARS-CoV-2–recovered nondialysis controls and significantly higher than virus-naive patients on dialysis and nondialysis controls (Figure 1C ). These levels after two doses were statistically higher in patients with prolonged delay between infection and first vaccination (>4 months) (Supplemental Figure 3 ). Age was associated with a reduced memory B cell expansion in naive patients on dialysis but not in SARS-CoV-2–recovered patients (Supplemental Figures 3 and 4 ). Moreover, after the third dose, no further increase of RBD-specific memory B cells was observed in SARS-CoV-2–recovered patients on dialysis. Furthermore, RBD-specific memory B cell frequencies were similar in SARS-CoV-2–recovered and virus-naive controls (Figure 1, C and D ). Lastly, we took advantage of serial samples in selected patients to show that frequencies of RBD-specific memory B cells remain stable in SARS-CoV-2–recovered patients on dialysis up to 6 months after the second dose. We conclude that the third vaccine dose (administered within 6 months after the second dose) induced an expansion of RBD-specific memory B cells in virus-naive patients on dialysis but had no significant effect on RBD-specific memory B cells in SARS-CoV-2–recovered patients on dialysis.
Finally, to better characterize the generated anti-RBD–specific B cell subsets in patients on dialysis, we performed multiparametric FACS analysis using a flow panel that included seven surface markers (CD19, CD21, CD27, CD11c+ , CD38, CD71, IgD) (Supplemental Figure 2, B and C ). The RBD-specific double-negative IgD− CD27− population exhibits an expansion in virus-naive patients on dialysis after the third dose (Figure 2A ), albeit with no difference in the repartition of atypical memory B cells (DN2; IgD− CD27+/− CD11c+ ) and antibody-secreting cells precursors (DN1; IgD− CD27− ) (Figure 2B ). No further increase of RBD-specific double-negative IgD− CD27 was observed in SARS-CoV-2–recovered patients on dialysis between the second and third doses (Figure 2B ).
Discussion
In this study, we describe antibody and memory B cell response after a third dose of mRNA vaccine in infection-naive and recovered patients on dialysis. Previous reports have suggested that the humoral response was impaired in patients on dialysis (9 ,15 ,22 ). We provide here a more complex view showing that virus-naive and SARS-CoV-2–recovered patients on dialysis display different profiles of immune response. Notably, a third dose of mRNA vaccine administered within 6 months after the second dose strongly boosted both serologic and cellular memory responses in virus-naive patients on dialysis. However, this boost was not observed in SARS-CoV-2–recovered patients previously vaccinated with two doses. As previously reported in larger studies (8 ,9 ), we show that the anti-RBD IgG response remained quantitatively lower (but nonsignificantly in our cohort) in virus-naive patients on dialysis compared with virus-naive healthy controls after two doses of mRNA vaccine. We demonstrate for the first time that RBD-specific memory B cells appeared settled at similar levels after two doses of mRNA vaccine in virus-naive patients on dialysis and nondialysis controls. Furthermore, these memory B cells were efficiently recalled by a third dose of mRNA vaccine as illustrated by the strong anti-RBD antibody response, exceeding the postsecond dose response in controls, and the high memory B cell frequencies, similar to those of SARS-CoV-2–recovered patients on dialysis. Of note, regarding the current third dose policy proposed to the general population, our data suggest a high benefit of this strategy in virus-naive individuals because this recall could be even more pronounced in the nondialysis population (15 ). Conversely, in SARS-CoV-2–recovered patients on dialysis, the third dose administered within 6 months did not further increase the anti-RBD IgG titers or the number of RBD-specific memory B cells , suggesting a limited benefit of such a rapid third dose in previously infected patients. Consequently, our results suggest that virus-naive patients on dialysis do not have a maximal immune response (antibodies as well as memory B cells ) after two doses, and therefore, a third dose can have a booster effect. Conversely, in SARS-CoV-2–recovered patients, the immune response is already maximally activated after infection and two doses, and therefore, a third dose given within 6 months may have no additional booster effect. However, a potential benefit of a delayed third dose in SARS-CoV-2–recovered patients and whether the same results would be found if the third dose was given later remain to be studied. In line with this hypothesis, a better response after two doses was observed in patients with a prolonged interval between infection and vaccination (Supplemental Figure 3 ). Moreover, even in the absence of an anti-S increase, we cannot rule out a benefit of the third dose in terms of antibody affinity and neutralizing potential. Lastly, how the memory B cell pool generated after vaccination will evolve remains to be studied.
In addition to serum IgG, memory B cells constitute another layer of immune protection. We and others have shown that mRNA vaccination generated a broad and diverse repertoire of RBD-specific memory B cells that can recognize SARS-CoV-2 variants of concern (13 ). In this study, we were able to characterize this response in patients on dialysis up to 6 months after vaccination. We found that patients on dialysis (either SARS-CoV-2 naive or recovered) achieve a quantitatively normal RBD memory B cell response after two or three doses of mRNA vaccine in SARS-CoV-2–recovered and virus-naive patients, respectively. Moreover, we took advantage of a prolonged longitudinal follow-up to show that RBD memory B cell response remains stable up to 6 months after the second dose in SARS-CoV-2–recovered patients on dialysis, as recently reported in SARS-CoV-2–recovered patients not on dialysis (23 ). Consequently, our data suggest that despite their immunocompromised status, patients on dialysis could settle a memory B cell response close to that of the general population. Whether the underlying repertoire of these RBD-specific memory B cells of patients on dialysis is as diverse and broadly neutralizing as in vaccinated healthy individuals remains an open question.
The current Omicron variant of concern is associated with very high transmissibility. From an epidemiologic point of view, our data could have major consequences in implementing future vaccination campaigns because the frequency of previous SARS-CoV-2 infection is obviously bound to increase in the dialysis population. By including the delay with (1 ) previous SARS-CoV-2 infection and (2 ) second dose in prioritization decisions, our results will lead to a better stratification of future vaccine policies. Notably, even though patients on dialysis seem to establish a memory B cell response similar to that of the general population, the characterization of the postvaccine B cell response in the other immunocompromised populations still needs to be studied.
The Omicron variant has become dominant in most countries. Omicron RBD harbors 15 mutations compared with the ancestral Hu-1 RBD analyzed in this study, and studies have shown decreased sera neutralization against this variant of concern. Although this limits the interpretation of our work, we recently showed that despite the strong immune escape of Omicron, the memory B cell pool generated after two doses of mRNA vaccines contains clones that maintained their neutralization potency against Omicron (24 ). This is in line with numerous reports showing increased neutralizing antibodies titers against Omicron after a third dose of mRNA vaccine, reflecting the recruitment of these memory B cells (25 ,26 ).
Our study has several limitations. Obviously, controls and patients on dialysis have different characteristics, which limit their comparison and suggest that dialysis status may not solely explain differences in immune responses. The relatively small size and the limited follow-up of the cohorts do not allow us to evaluate the role of comorbidities (such as immunosuppression) in patients on dialysis or predict long-term response to mRNA vaccine and its persistence, although our results precisely characterize memory B cell response up to 6 months after vaccination. Also, longitudinal follow-up was not available for all patients. Given the retrospective design of the study, different timings between the second and third doses and SARS-CoV-2 infection and vaccination were possible. However, similarly to our whole cohort, we were able to show in selected patients with serial samples the increase of memory B cell response in virus-naive patients and the stability of this response in SARS-CoV-2–recovered patients (even without a third dose) (Figure 1, C and D ). Moreover, our analysis in infection-experienced patients on dialysis may be altered by survivor bias, limiting the generalization of our observations to other at-risk populations. We cannot rule out that there may have been misclassification of previously infected status, at least of asymptomatic cases. Lastly, we evaluated responses to only BNT162B2 and not to other vaccines (including other mRNA vaccines or other vaccine platforms).
In conclusion, our study shows that previous SARS-CoV-2 infection strongly affects vaccine responses in patients on dialysis. The third dose of mRNA vaccine administered within 6 months after the second dose boosts the serologic and memory response in virus-naive patients on dialysis, but this boost was not observed in SARS-CoV-2–recovered patients on dialysis. These results suggest that the timing of the third dose and a previous SARS-CoV-2 infection have a critical effect in vaccine response of patients on dialysis and should be considered in future vaccine policies of this population.
Disclosures
K. El Karoui reports research funding from Amgen, Otsuka, and Sanofi and honoraria from Alexion, AstraZeneca, and Otsuka. S. Fourati reports consultancy agreements with Abbott Diagnostics, GeneXpert, GlaxoSmithKline (GSK), and MSD; honoraria from Abbott Diagnostics, GeneXpert, GSK, and MSD; serving in an advisory or leadership role for Abbott Diagnostics, GeneXpert, GSK, and MSD; and speakers bureau for Abbott Diagnostics, GeneXpert, GSK, and MSD. M. Mahévas reports research funding from GSK and Takeda and honoraria from Amgen and Novartis. All remaining authors have nothing to disclose.
Funding
None.
Acknowledgments
We thank Dr. C.A. Reynaud and Dr. J.C. Weill for their helpful comments.
Author Contributions
P. Attias, K. El Karoui, M. Mahévas, and H. Sakhi conceptualized the study; P. Attias, I. Azzaoui, M. Bouvier, A. de La Selle, H. Sakhi, and A. Sokal were responsible for data curation; P. Attias, D. Dahmane, K. El Karoui, P. Nizard, and P. Rieu were responsible for investigation; I. Azzaoui, A. de La Selle, K. El Karoui, S. Fourati, M. Mahévas, H. Sakhi, and A. Sokal were responsible for formal analysis; P. Attias, I. Azzaoui, K. El Karoui, S. Fourati, M. Mahévas, H. Sakhi, and A. Sokal wrote the original draft; and P. Attias, I. Azzaoui, M. Bouvier, P. Chappert, D. Dahmane, A. de La Selle, K. El Karoui, I. Fernandez, S. Fourati, P. Grimbert, M. Mahévas, P. Nizard, P. Rieu, H. Sakhi, C. Samson, and A. Sokal reviewed and edited the manuscript.
Data Sharing Statement
All data used in this study are available in this article.
Supplemental Material
This article contains supplemental material online at http://cjasn.asnjournals.org/lookup/suppl/doi:10.2215/CJN.00830122/-/DCSupplemental .
Supplemental Materials . Methods.
Supplemental Table 1 . Conjugated antibodies used in flow cytometry.
Supplemental Table 2 . Median values of anti–SARS-CoV-2 IgG titer and frequency of SARS-CoV-2 RBD-specific memory B cells (percentage of CD27+ IgD− ) in virus-naive healthy controls and patients on dialysis after two doses and three doses.
Supplemental Table 3 . Median values of anti–SARS-CoV-2 IgG titer and frequency of SARS-CoV-2 RBD-specific memory B cells (percentage of CD27+ IgD− ) in SARS-CoV-2–recovered healthy controls and patients on dialysis after two doses and three doses.
Supplemental Table 4 . Baseline characteristics of naive patients on hemodialysis according to the number of vaccine doses.
Supplemental Table 5 . Baseline characteristics of SARS-CoV-2–recovered patients on hemodialysis according to the number of vaccine doses.
Supplemental Figure 1 . Cohort design.
Supplemental Figure 2 . Gating strategies.
Supplemental Figure 3 . Humoral and memory B cell response to SARS-CoV-2 RBD in SARS-CoV-2–recovered patients on dialysis after two mRNA vaccine doses according to baseline characteristics.
Supplemental Figure 4 . Humoral and memory B cell response to SARS-CoV-2 RBD in SARS-CoV-2–naive patients on dialysis after two mRNA vaccine doses according to baseline characteristics.
References
1. Jager KJ, Kramer A, Chesnaye NC, Couchoud C, Sánchez-Álvarez JE, Garneata L, Collart F, Hemmelder MH, Ambühl P, Kerschbaum J, Legeai C, Del Pino Y Pino MD, Mircescu G, Mazzoleni L, Hoekstra T, Winzeler R, Mayer G, Stel VS, Wanner C, Zoccali C, Massy ZA: Results from the ERA-EDTA Registry indicate a high mortality due to COVID-19 in dialysis patients and kidney transplant recipients across Europe. Kidney Int 98: 1540–1548, 2020
2. Couchoud C, Bayer F, Ayav C, Béchade C, Brunet P, Chantrel F, Frimat L, Galland R, Hourmant M, Laurain E, Lobbedez T, Mercadal L, Moranne O; French REIN registry: Low incidence of SARS-CoV-2, risk factors of mortality and the course of illness in the French national cohort of dialysis patients. Kidney Int 98: 1519–1529, 2020
3. Williamson EJ, Walker AJ, Bhaskaran K, Bacon S, Bates C, Morton CE, Curtis HJ, Mehrkar A, Evans D, Inglesby P, Cockburn J, McDonald HI, MacKenna B, Tomlinson L, Douglas IJ, Rentsch CT, Mathur R, Wong AYS, Grieve R, Harrison D, Forbes H, Schultze A, Croker R, Parry J, Hester F, Harper S, Perera R, Evans SJW, Smeeth L, Goldacre B: Factors associated with COVID-19-related death using OpenSAFELY. Nature 584: 430–436, 2020
4. De Meester J, De Bacquer D, Naesens M, Meijers B, Couttenye MM, De Vriese AS; NBVN Kidney Registry Group: Incidence, characteristics, and outcome of COVID-19 in adults on kidney replacement therapy: A regionwide registry study. J Am Soc Nephrol 32: 385–396, 2021
5. Clarke C, Prendecki M, Dhutia A, Ali MA, Sajjad H, Shivakumar O, Lightstone L, Kelleher P, Pickering MC, Thomas D, Charif R, Griffith M, McAdoo SP, Willicombe M: High prevalence of asymptomatic COVID-19 infection in hemodialysis patients detected using serologic screening. J Am Soc Nephrol 31: 1969–1975, 2020
6. Dagan N, Barda N, Kepten E, Miron O, Perchik S, Katz MA, Hernán MA, Lipsitch M, Reis B, Balicer RD: BNT162b2 mRNA Covid-19 vaccine in a nationwide mass vaccination setting. N Engl J Med 384: 1412–1423, 2021
7. Ikizler TA, Coates PT, Rovin BH, Ronco P: Immune response to SARS-CoV-2 infection and vaccination in patients receiving kidney replacement therapy. Kidney Int 99: 1275–1279, 2021
8. Espi M, Charmetant X, Barba T, Koppe L, Pelletier C, Kalbacher E, Chalencon E, Mathias V, Ovize A, Cart-Tanneur E, Bouz C, Pellegrina L, Morelon E, Fouque D, Juillard L, Thaunat O: The ROMANOV study found impaired humoral and cellular immune responses to SARS-CoV-2 mRNA vaccine in virus-unexposed patients receiving maintenance hemodialysis. Kidney Int 100: 928–936, 2021
9. Van Praet J, Reynders M, De Bacquer D, Viaene L, Schoutteten M, Caluwé R, Doubel P, Heylen L, De Bel AV, Van Vlem B, Steensels D, De Vriese AS: Predictors and dynamics of the humoral and cellular immune response to SARS-CoV-2 mRNA vaccines in hemodialysis patients: A multicenter observational study. J Am Soc Nephrol 32: 3208–3220, 2021
10. Stumpf J, Siepmann T, Lindner T, Karger C, Schwöbel J, Anders L, Faulhaber-Walter R, Schewe J, Martin H, Schirutschke H, Barnett K, Hüther J, Müller P, Langer T, Pluntke T, Anding-Rost K, Meistring F, Stehr T, Pietzonka A, Escher K, Cerny S, Rothe H, Pistrosch F, Seidel H, Paliege A, Beige J, Bast I, Steglich A, Gembardt F, Kessel F, Kröger H, Arndt P, Sradnick J, Frank K, Klimova A, Mauer R, Grählert X, Anft M, Blazquez-Navarro A, Westhoff TH, Stervbo U, Tonn T, Babel N, Hugo C: Humoral and cellular immunity to SARS-CoV-2 vaccination in renal transplant versus dialysis patients: A prospective, multicenter observational study using mRNA-1273 or BNT162b2 mRNA vaccine. Lancet Reg Health Eur 9: 100178, 2021
11. Sibbel S, McKeon K, Luo J, Wendt K, Walker AG, Kelley T, Lazar R, Zywno ML, Connaire JJ, Tentori F, Young A, Brunelli SM: Real-world effectiveness and immunogenicity of BNT162b2 and mRNA-1273 SARS-CoV-2 vaccines in patients on hemodialysis. J Am Soc Nephrol 33: 49–57, 2022
12. Espi M, Charmetant X, Barba T, Mathieu C, Pelletier C, Koppe L, Chalencon E, Kalbacher E, Mathias V, Ovize A, Cart-Tanneur E, Bouz C, Pellegrina L, Morelon E, Juillard L, Fouque D, Couchoud C, Thaunat O; REIN Registry: A prospective observational study for justification, safety, and efficacy of a third dose of mRNA vaccine in patients receiving maintenance hemodialysis. Kidney Int 100: 928–936, 2021
13. Cho A, Muecksch F, Schaefer-Babajew D, Wang Z, Finkin S, Gaebler C, Ramos V, Cipolla M, Mendoza P, Agudelo M, Bednarski E, DaSilva J, Shimeliovich I, Dizon J, Daga M, Millard KG, Turroja M, Schmidt F, Zhang F, Tanfous TB, Jankovic M, Oliveria TY, Gazumyan A, Caskey M, Bieniasz PD, Hatziioannou T, Nussenzweig MC: Anti-SARS-CoV-2 receptor-binding domain antibody evolution after mRNA vaccination. Nature 600: 517–522, 2021
14. Sokal A, Barba-Spaeth G, Fernández I, Broketa M, Azzaoui I, de La Selle A, Vandenberghe A, Fourati S, Roeser A, Meola A, Bouvier-Alias M, Crickx E, Languille L, Michel M, Godeau B, Gallien S, Melica G, Nguyen Y, Zarrouk V, Canoui-Poitrine F, Pirenne F, Mégret J, Pawlotsky JM, Fillatreau S, Bruhns P, Rey FA, Weill JC, Reynaud CA, Chappert P, Mahévas M: mRNA vaccination of naive and COVID-19-recovered individuals elicits potent
memory B cells that recognize SARS-CoV-2 variants. Immunity 54: 2893–2907.e5, 2021
15. Rincon-Arevalo H, Choi M, Stefanski AL, Halleck F, Weber U, Szelinski F, Jahrsdörfer B, Schrezenmeier H, Ludwig C, Sattler A, Kotsch K, Potekhin A, Chen Y, Burmester GR, Eckardt KU, Guerra GM, Durek P, Heinrich F, Ferreira-Gomes M, Radbruch A, Budde K, Lino AC, Mashreghi MF, Schrezenmeier E, Dörner T: Impaired humoral immunity to SARS-CoV-2 BNT162b2 vaccine in kidney transplant recipients and dialysis patients. Sci Immunol 6: eabj1031, 2021
16. Kato S, Chmielewski M, Honda H, Pecoits-Filho R, Matsuo S, Yuzawa Y, Tranaeus A, Stenvinkel P, Lindholm B: Aspects of immune dysfunction in end-stage renal disease. Clin J Am Soc Nephrol 3: 1526–1533, 2008
17. Ducloux D, Colladant M, Chabannes M, Yannaraki M, Courivaud C: Humoral response after 3 doses of the BNT162b2 mRNA COVID-19 vaccine in patients on hemodialysis. Kidney Int 100: 702–704, 2021
18. Longlune N, Nogier MB, Miedougé M, Gabilan C, Cartou C, Seigneuric B, Del Bello A, Marion O, Faguer S, Izopet J, Kamar N: High immunogenicity of a messenger RNA-based vaccine against SARS-CoV-2 in chronic dialysis patients. Nephrol Dial Transplant 36: 1704–1709, 2021
19. Stervbo U, Blazquez-Navarro A, Blanco EV, Safi L, Meister TL, Paniskaki K, Stockhausen M, Marheinecke C, Zimmer G, Wellenkötter J, Giglio T, Arora P, Pöhlmann S, Hoffmann M, Seibert FS, Pfaender S, Roch T, Westhoff TH, Cinkilic O, Babel N: Improved cellular and humoral immunity upon a second BNT162b2 and mRNA-1273 boost in prime-boost vaccination no/low responders with end-stage renal disease. Kidney Int 100: 1335–1337, 2021
20. Anand S, Montez-Rath M, Han J, Cadden L, Hunsader P, Kerschmann R, Beyer P, Boyd SD, Garcia P, Dittrich M, Block GA, Parsonnet J, Chertow GM: Estimated SARS-CoV-2 seroprevalence in US patients receiving dialysis 1 year after the beginning of the COVID-19 pandemic. JAMA Netw Open 4: e2116572, 2021
21. Sokal A, Barba-Spaeth G, Fernández I, Broketa M, Azzaoui I, de La Selle A, Vandenberghe A, Fourati S, Roeser A, Meola A, Bouvier-Alias M, Crickx E, Languille L, Michel M, Godeau B, Gallien S, Melica G, Nguyen Y, Zarrouk V, Canoui-Poitrine F, Pirenne F, Mégret J, Pawlotsky J-M, Fillatreau S, Bruhns P, Rey FA, Weill J-C, Reynaud C-A, Chappert P, Mahévas M: mRNA vaccination of naive and COVID-19-recovered individuals elicits potent
memory B cells that recognize SARS-CoV-2 variants. Immunity 54: 2893–2907, 2021
22. Sattler A, Schrezenmeier E, Weber UA, Potekhin A, Bachmann F, Straub-Hohenbleicher H, Budde K, Storz E, Proß V, Bergmann Y, Thole LM, Tizian C, Hölsken O, Diefenbach A, Schrezenmeier H, Jahrsdörfer B, Zemojtel T, Jechow K, Conrad C, Lukassen S, Stauch D, Lachmann N, Choi M, Halleck F, Kotsch K: Impaired humoral and cellular immunity after SARS-CoV-2 BNT162b2 (tozinameran) prime-boost vaccination in kidney transplant recipients. J Clin Invest 131: 150175, 2021
23. Goel RR, Painter MM, Apostolidis SA, Mathew D, Meng W, Rosenfeld AM, Lundgreen KA, Reynaldi A, Khoury DS, Pattekar A, Gouma S, Kuri-Cervantes L, Hicks P, Dysinger S, Hicks A, Sharma H, Herring S, Korte S, Baxter AE, Oldridge DA, Giles JR, Weirick ME, McAllister CM, Awofolaju M, Tanenbaum N, Drapeau EM, Dougherty J, Long S, D’Andrea K, Hamilton JT, McLaughlin M, Williams JC, Adamski S, Kuthuru O, Frank I, Betts MR, Vella LA, Grifoni A, Weiskopf D, Sette A, Hensley SE, Davenport MP, Bates P, Luning Prak ET, Greenplate AR, Wherry EJ; UPenn COVID Processing Unit: mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science 374: abm0829, 2021
24. Sokal A, Broketa M, Barba-Spaeth G, Meola A, Fernández I, Fourati S, Azzaoui I, La Selle A, Vandenberghe A, Roeser A, Bouvier-Alias M, Crickx E, Languille L, Michel M, Godeau B, Gallien S, Melica G, Nguyen Y, Zarrouk V, Canoui-Poitrine F, Noizat-Pirenne F, Megret J, Pawlotsky J-M, Fillatreau S, Simon-Lorière E, Weill J-C, Reynaud C-A, Rey FA, Bruhns P, Chappert P, Mahévas M: Analysis of mRNA vaccination-elicited RBD-specific
memory B cells reveals strong but incomplete immune escape of the SARS-CoV-2 Omicron variant [published online ahead of print April 7, 2022]. Immunity 10.1016/j.immuni.2022.04.002
25. Gruell H, Vanshylla K, Tober-Lau P, Hillus D, Schommers P, Lehmann C, Kurth F, Sander LE, Klein F: mRNA booster immunization elicits potent neutralizing serum activity against the SARS-CoV-2 Omicron variant. Nat Med 28: 477–480, 2022
26. Garcia-Beltran WF, St Denis KJ, Hoelzemer A, Lam EC, Nitido AD, Sheehan ML, Berrios C, Ofoman O, Chang CC, Hauser BM, Feldman J, Roederer AL, Gregory DJ, Poznansky MC, Schmidt AG, Iafrate AJ, Naranbhai V, Balazs AB: mRNA-based COVID-19 vaccine boosters induce neutralizing immunity against SARS-CoV-2 Omicron variant. Cell 185: 457–466.e4, 2022