Humoral and cellular immune responses to two and three doses of SARS-CoV-2 vaccines in rituximab-treated patients with rheumatoid arthritis: a prospective, cohort study

Ingrid Jyssum, Hassen Kared, Trung T Tran, Anne T Tveter, Sella A Provan, Joseph Sexton, Kristin K Jørgensen, Jørgen Jahnsen, Grete B Kro, David J Warren, Eline B Vaage, Tore K Kvien, Lise-Sofie H Nissen-Meyer, Ane Marie Anderson, Gunnveig Grødeland, Espen A Haavardsholm, John Torgils Vaage, Siri Mjaaland, Silje Watterdal Syversen, Fridtjof Lund-Johansen, Ludvig A Munthe, Guro Løvik Goll, Ingrid Jyssum, Hassen Kared, Trung T Tran, Anne T Tveter, Sella A Provan, Joseph Sexton, Kristin K Jørgensen, Jørgen Jahnsen, Grete B Kro, David J Warren, Eline B Vaage, Tore K Kvien, Lise-Sofie H Nissen-Meyer, Ane Marie Anderson, Gunnveig Grødeland, Espen A Haavardsholm, John Torgils Vaage, Siri Mjaaland, Silje Watterdal Syversen, Fridtjof Lund-Johansen, Ludvig A Munthe, Guro Løvik Goll

Abstract

Background: In rituximab-treated patients with rheumatoid arthritis, humoral and cellular immune responses after two or three doses of SARS-CoV-2 vaccines are not well characterised. We aimed to address this knowledge gap.

Methods: This prospective, cohort study (Nor-vaC) was done at two hospitals in Norway. For this sub-study, we enrolled patients with rheumatoid arthritis on rituximab treatment and healthy controls who received SARS-CoV-2 vaccines according to the Norwegian national vaccination programme. Patients with insufficient serological responses to two doses (antibody to the receptor-binding domain [RBD] of the SARS-CoV-2 spike protein concentration <100 arbitrary units [AU]/mL) were allotted a third vaccine dose. Antibodies to the RBD of the SARS-CoV-2 spike protein were measured in serum 2-4 weeks after the second and third doses. Vaccine-elicited T-cell responses were assessed in vitro using blood samples taken before and 7-10 days after the second dose and 3 weeks after the third dose from a subset of patients by stimulating cryopreserved peripheral blood mononuclear cells with spike protein peptides. The main outcomes were the proportions of participants with serological responses (anti-RBD antibody concentrations of ≥70 AU/mL) and T-cell responses to spike peptides following two and three doses of SARS-CoV-2 vaccines. The study is registered at ClinicalTrials.gov, NCT04798625, and is ongoing.

Findings: Between Feb 9, 2021, and May 27, 2021, 90 patients were enrolled, 87 of whom donated serum and were included in our analyses (69 [79·3%] women and 18 [20·7%] men). 1114 healthy controls were included (854 [76·7%] women and 260 [23·3%] men). 49 patients were allotted a third vaccine dose. 19 (21·8%) of 87 patients, compared with 1096 (98·4%) of 1114 healthy controls, had a serological response after two doses (p<0·0001). Time since last rituximab infusion (median 267 days [IQR 222-324] in responders vs 107 days [80-152] in non-responders) and vaccine type (mRNA-1273 vs BNT162b2) were significantly associated with serological response (adjusting for age and sex). After two doses, 10 (53%) of 19 patients had CD4+ T-cell responses and 14 (74%) had CD8+ T-cell responses. A third vaccine dose induced serological responses in eight (16·3%) of 49 patients, but induced CD4+ and CD8+ T-cell responses in all patients assessed (n=12), including responses to the SARS-CoV-2 delta variant (B.1.617.2). Adverse events were reported in 32 (48%) of 67 patients and in 191 (78%) of 244 healthy controls after two doses, with the frequency not increasing after the third dose. There were no serious adverse events or deaths.

Interpretation: This study provides important insight into the divergent humoral and cellular responses to two and three doses of SARS-CoV-2 vaccines in rituximab-treated patients with rheumatoid arthritis. A third vaccine dose given 6-9 months after a rituximab infusion might not induce a serological response, but could be considered to boost the cellular immune response.

Funding: The Coalition for Epidemic Preparedness Innovations, Research Council of Norway Covid, the KG Jebsen Foundation, Oslo University Hospital, the University of Oslo, the South-Eastern Norway Regional Health Authority, Dr Trygve Gythfeldt og frues forskningsfond, the Karin Fossum Foundation, and the Research Foundation at Diakonhjemmet Hospital.

Conflict of interest statement

KKJ reports speakers bureaus from Roche and BMS and advisory board participation for Celltrion and Norgine. JJ reports grants from Abbvie, Pharmacosmos, and Ferring; consulting fees from Abbvie, Boerhinger Ingelheim, BMS, Celltrion, Ferring, Glihead, Janssen Cilag, MSD, Napp Pharma, Novartis, Orion Pharma, Pfeizer, Pharmacosmos, Takeda, Sandoz, and Unimedic Pharma; and speakers bureaus from Abbvie, Astro Pharma, Boerhinger Ingelheim, BMS, Celltrion, Ferring, Glihead, Hikma, Janssen Cilag, Meda, MSD, Napp Pharma, Novartis, Oriuon Pharma, Pfeizer, Pharmacosmos, Roche, Takeda, and Sandoz. TKK reports grants from AbbVie, Amgen, BMS, MSD, Novartis, Pfizer, and UCB; consulting fees from AbbVie, Amgen, Biogen, Celltrion, Eli Lilly, Gilead, Mylan, Novartis, Pfizer, Sandoz, and Sanofi; speakers bureaus from Amgen, Celltrion, Egis, Evapharma, Ewopharma, Hikma, Oktal, Sandoz, and Sanofi; and participation on a data safety monitoring board for AbbVie. LAM reports funding from the KG Jebsen foundation; support for infrastructure and biobanking from the University of Oslo and Oslo University Hospital; grants from the Coalition of Epidemic Preparedness Innovations (CEPI); and speakers bureaus from Novartis and Cellgene. GG reports consulting fees from the Norwegian System of Compensation to Patients and AstraZeneca, and speakers bureaus from Bayer, Sanofi Pasteur, and Thermo Fisher. JTV reports grant from the CEPI. FL-J reports grants from the CEPI and the South-Eastern Norway Regional Health Authority. GLG reports funding from the Karin Fossum foundation, Diakonhjemmet Hospital, Oslo University Hospital, Akershus University Hospital, the Dr Trygve Gydtfeldt og frues Foundation, and the South-Eastern Norway Regional Health Authority; consulting fees from AbbVie and Pfizer; speakers fees from AbbVie, Pfizer, Sandoz, Orion Pharma, Novartis, and UCB; and advisory board participation for Pfizer and AbbVie. All other authors declare no competing interests.

© 2021 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Humoral response to two and three vaccine doses (A) Anti-RBD antibody concentrations in controls, patients who had received at least two doses, patients who had received two doses and would later receive a third, and patients who had received three doses. The violin illustrates the kernel probability density and the orange line indicates the median. Dots denote individual patients. (B) Time between last rituximab infusion and first vaccine dose according to response status in all patients after their second vaccine dose. The violin illustrates the kernel probability density and the orange line indicates the median. Dots denote individual patients. (C) Anti-RBD antibody concentrations after the second and third doses. Solid lines connect patients' two samples (circles). The horizontal dotted line indicates the cutoff for positivity (70 AU/mL). (D) Time between the last rituximab infusion and anti-RBD response after the third vaccine dose. AU=arbitrary units. RBD=receptor-binding domain.
Figure 2
Figure 2
T-cell responses after two and three vaccine doses (A) Anti-wild-type spike protein-specific T-cell responses in patients after two and three doses and in healthy controls after two doses. CD4+ T-cell responses and CD8+ T-cell responses are shown for all unstimulated and stimulated pairs. The p values from Wilcoxon matched pairs signed rank tests are shown, with * indicating p<0·001 and †indicating p<0·0001. Patients with a response (positive) and patients without a response (negative) are indicated. (B) Analysis of T-cell responses directed against wild-type and delta variant SARS-CoV-2 spike peptides in patients after two and three doses (Spearman correlation). Solid lines show simple linear regression of correlation and dotted lines represent 95% CIs. (C) Percentage of anti-spike protein CD4+ T cells versus anti-spike protein CD8+ T cells in patient responders to wild-type and delta variant spike peptides using combined data of the second and third doses. Spearman correlation is shown. (D) Percentage of anti-spike protein CD4+ T cells versus age in patient responders to wild-type and delta variant spike peptides using combined data of the second and third doses. Spearman correlation is shown. See the appendix (p 5) for supplementary data for gating and controls.
Figure 3
Figure 3
Adverse events following two or three vaccine doses in patients and controls (A) All patients. (B) Controls. (C) Patients who received three vaccine doses. Adverse events were reported for all patients and a subset (n=246) of healthy controls (health-care workers at Diakonhjemmet Hospital and Akershus University Hospital, Oslo, Norway). *Duration not measured. †No patients were hospitalised due to disease flares after vaccination.

References

    1. Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020;383:2603–2615.
    1. Baden LR, El Sahly HM, Essink B, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2021;384:403–416.
    1. Friedman MA, Curtis JR, Winthrop KL. Impact of disease-modifying antirheumatic drugs on vaccine immunogenicity in patients with inflammatory rheumatic and musculoskeletal diseases. Ann Rheum Dis. 2021;80:1255–1265.
    1. Avouac J, Drumez E, Hachulla E, et al. COVID-19 outcomes in patients with inflammatory rheumatic and musculoskeletal diseases treated with rituximab: a cohort study. Lancet Rheumatol. 2021;3:e419–e426.
    1. Fagni F, Simon D, Tascilar K, et al. COVID-19 and immune-mediated inflammatory diseases: effect of disease and treatment on COVID-19 outcomes and vaccine responses. Lancet Rheumatol. 2021;3:e724–e736.
    1. Raiker R, DeYoung C, Pakhchanian H, et al. Outcomes of COVID-19 in patients with rheumatoid arthritis: a multicenter research network study in the United States. Semin Arthritis Rheum. 2021;51:1057–1066.
    1. Andersen KM, Bates BA, Rashidi ES, et al. Long-term use of immunosuppressive medicines and in-hospital COVID-19 outcomes: a retrospective cohort study using data from the National COVID Cohort Collaborative. Lancet Rheumatol. 2021 doi: 10.1016/S2665-9913(21)00325-8. published online Nov 15.
    1. Jena A, Mishra S, Deepak P, et al. Response to SARS-CoV-2 vaccination in immune mediated inflammatory diseases: systematic review and meta-analysis. Autoimmun Rev. 2021 doi: 10.1016/j.autrev.2021.102927. published online Aug 30.
    1. Mrak D, Tobudic S, Koblischke M, et al. SARS-CoV-2 vaccination in rituximab-treated patients: B cells promote humoral immune responses in the presence of T-cell-mediated immunity. Ann Rheum Dis. 2021;80:1345–1350.
    1. Furer V, Eviatar T, Zisman D, et al. Immunogenicity and safety of the BNT162b2 mRNA COVID-19 vaccine in adult patients with autoimmune inflammatory rheumatic diseases and in the general population: a multicentre study. Ann Rheum Dis. 2021;80:1330–1338.
    1. Moor MB, Suter-Riniker F, Horn MP, et al. Humoral and cellular responses to mRNA vaccines against SARS-CoV-2 in patients with a history of CD20 B-cell-depleting therapy (RituxiVac): an investigator-initiated, single-centre, open-label study. Lancet Rheumatol. 2021;3:e789–e797.
    1. McMahan K, Yu J, Mercado NB, et al. Correlates of protection against SARS-CoV-2 in rhesus macaques. Nature. 2021;590:630–634.
    1. Rydyznski Moderbacher C, Ramirez SI, Dan JM, et al. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell. 2020;183:996–1012.e19.
    1. Bange EM, Han NA, Wileyto P, et al. CD8+ T cells contribute to survival in patients with COVID-19 and hematologic cancer. Nat Med. 2021;27:1280–1289.
    1. Bonelli MM, Mrak D, Perkmann T, Haslacher H, Aletaha D. SARS-CoV-2 vaccination in rituximab-treated patients: evidence for impaired humoral but inducible cellular immune response. Ann Rheum Dis. 2021;80:1355–1356.
    1. Alexander JL, Selinger CP, Powell N. Third doses of SARS-CoV-2 vaccines in immunosuppressed patients with inflammatory bowel disease. Lancet Gastroenterol Hepatol. 2021;6:987–988.
    1. Bar-On YM, Goldberg Y, Mandel M, et al. Protection of BNT162b2 vaccine booster against Covid-19 in Israel. N Engl J Med. 2021;385:1393–1400.
    1. Felten R, Gallais F, Schleiss C, et al. Cellular and humoral immunity after the third dose of SARS-CoV-2 vaccine in patients treated with rituximab. Lancet Rheumatol. 2021 doi: 10.1016/S2665-9913(21)00351-9. published online Nov 8.
    1. Norwegian Institute of Public Health Norwegian Immunisation Registry SYSVAK.
    1. Norwegian Institute of Public Health Norwegian Surveillance System for Communicable Diseases (MSIS)
    1. Holter JC, Pischke SE, de Boer E, et al. Systemic complement activation is associated with respiratory failure in COVID-19 hospitalized patients. Proc Natl Acad Sci USA. 2020;117:25018–25025.
    1. König M, Lorentzen ÅR, Torgauten HM, et al. Humoral immunity to SARS-CoV-2 mRNA vaccination in multiple sclerosis: the relevance of time since last rituximab infusion and first experience from sporadic revaccinations. J Neurol Neurosurg Psychiatry. 2021 doi: 10.1136/jnnp-2021-327612. published online Oct 20.
    1. Khoury DS, Cromer D, Reynaldi A, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med. 2021;27:1205–1211.
    1. Bergwerk M, Gonen T, Lustig Y, et al. Covid-19 breakthrough infections in vaccinated health care workers. N Engl J Med. 2021;385:1474–1484.
    1. Chia WN, Zhu F, Ong SWX, et al. Dynamics of SARS-CoV-2 neutralising antibody responses and duration of immunity: a longitudinal study. Lancet Microbe. 2021;2:e240–e249.
    1. Le Bert N, Tan AT, Kunasegaran K, et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature. 2020;584:457–462.
    1. Steensels D, Pierlet N, Penders J, Mesotten D, Heylen L. Comparison of SARS-CoV-2 antibody response following vaccination with BNT162b2 and mRNA-1273. JAMA. 2021;326:1533–1535.
    1. Collier DA, Ferreira IATM, Kotagiri P, et al. Age-related immune response heterogeneity to SARS-CoV-2 vaccine BNT162b2. Nature. 2021;596:417–422.
    1. Richards NE, Keshavarz B, Workman LJ, Nelson MR, Platts-Mills TAE, Wilson JM. Comparison of SARS-CoV-2 antibody response by age among recipients of the BNT162b2 vs the mRNA-1273 vaccine. JAMA Netw Open. 2021;4

Source: PubMed

3
Tilaa