COVID-19 vaccine-readiness for anti-CD20-depleting therapy in autoimmune diseases

D Baker, C A K Roberts, G Pryce, A S Kang, M Marta, S Reyes, K Schmierer, G Giovannoni, S Amor, D Baker, C A K Roberts, G Pryce, A S Kang, M Marta, S Reyes, K Schmierer, G Giovannoni, S Amor

Abstract

Although most autoimmune diseases are considered to be CD4 T cell- or antibody-mediated, many respond to CD20-depleting antibodies that have limited influence on CD4 and plasma cells. This includes rituximab, oblinutuzumab and ofatumumab that are used in cancer, rheumatoid arthritis and off-label in a large number of other autoimmunities and ocrelizumab in multiple sclerosis. Recently, the COVID-19 pandemic created concerns about immunosuppression in autoimmunity, leading to cessation or a delay in immunotherapy treatments. However, based on the known and emerging biology of autoimmunity and COVID-19, it was hypothesised that while B cell depletion should not necessarily expose people to severe SARS-CoV-2-related issues, it may inhibit protective immunity following infection and vaccination. As such, drug-induced B cell subset inhibition, that controls at least some autoimmunities, would not influence innate and CD8 T cell responses, which are central to SARS-CoV-2 elimination, nor the hypercoagulation and innate inflammation causing severe morbidity. This is supported clinically, as the majority of SARS-CoV-2-infected, CD20-depleted people with autoimmunity have recovered. However, protective neutralizing antibody and vaccination responses are predicted to be blunted until naive B cells repopulate, based on B cell repopulation kinetics and vaccination responses, from published rituximab and unpublished ocrelizumab (NCT00676715, NCT02545868) trial data, shown here. This suggests that it may be possible to undertake dose interruption to maintain inflammatory disease control, while allowing effective vaccination against SARS-CoV-29, if and when an effective vaccine is available.

Keywords: B cell; CD20; COVID-19; autoimmunity; immunotherapy; multiple sclerosis; ocrelizumab; rheumatoid arthritis; rituximab.

Conflict of interest statement

D. B., M. M., K. S. and G. G. have received compensation for either consultancies and presentations and advisory board activities from Genentech/Roche. However, Roche/Genentech were not involved in the decision to write and submit this manuscript. S. A. has received consultancy from Novartis and Roche. C. A. K. K., G. P., A. S. K. and S. R. have nothing to disclose. D. B. has received compensation for activities related to Canbex therapeutics, InMune Biol, Lundbeck, Japan tobacco, Merck and Novartis. M. M. has received speaking honoraria from Sanofi‐Genzyme. K. S. has received compensation for activities related to Biogen, Eisai, Elan, Fiveprime, Lipomed, Merck KGAa, Novartis, Sanofi‐Genzyme and Teva. G. G. has received compensation for activities from AbbVie, Actelion, Atara Bio, Bayer‐Schering Healthcare, Biogen, Celgene, GW Pharma, GSK, Ironwood, Japanese Tobacco, Merck, Merck‐Serono, Mertz, Novartis, Pfizer, Sanofi‐Genzyme, Synthon, Takeda, Teva, UCB Pharma and Vertex Pharmaceuticals.

© 2020 The Authors. Clinical & Experimental Immunology published by John Wiley & Sons Ltd on behalf of British Society for Immunology.

Figures

Fig. 1
Fig. 1
Pathobiology of COVID‐19. Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infects cells in the lung and the gut via the angiotensin‐converting enzyme 2 (ACE2). This blocks ACE2‐induced formation of anti‐oxidant angiotensin, facilitating oxygen free‐radical formation and vascular damage. The innate immune response provides the initial line of defence against the virus, while a CD8 anti‐viral T cell response and neutralizing and complement‐fixing antibody response serve to remove the virus in the majority of people. However, the virus triggers suppression of interferon responses and other viral escape mechanisms that in a minority of people stimulate the innate immune response leading to lymphocyte apoptosis that blocks their regulatory signals and, in some cases, releases a cytokine storm that drives hyper‐innate inflammation. This, in part, causes acute respiratory distress. Importantly, this augments vascular damage that accentuates the respiratory distress and leads to von Willebrand factor release into the blood. This contributes to the formation of microthrombi, contributing to respiratory distress and vascular embolism that may be fatal. Adapted from Henry et al. 2020 [22].
Fig. 2
Fig. 2
Ocrelizumab inhibits vaccination responses. People with multiple sclerosis who did not receive ocrelizumab (control) or were infused with 300 mg ocrelizumab on days 0 and 15 and were vaccinated from weeks 12–24 after ocrelizumab. The experimental details and results were from www.clinicaltrials.gov NCT02545868 [91]. The results show: (a) The frequency of seroconversion in people treated with ocrelizumab following injection pneumococcal 23‐polyvalent pneumococcal vaccine (PPV) vaccine, 4 weeks after vaccination (n = 66–68). A 23‐PPV vaccine response against a serotype was defined by a twofold increase in anti‐pneumococcal antibody or > 1 µg/ml compared with prevaccination levels, following Food and Drug Administration guidance. (b) The titre of response to the initial challenge with 23‐PPV 4 weeks after vaccination. (c) The frequency of seroconversion in people treated with ocrelizumab following injection of a booster pneumococcal 13‐PPV vaccine 4 weeks after 23‐PPV (n = 33–34). The frequency of responders is shown 8 weeks after 23‐PPV vaccination. (d) The geometric mean and 95% confidence interval (CI) anti‐tetanus toxoid antibody levels measured by enzyme‐linked immunosorbent assay (ELISA) before and following vaccination (n = 34–68). (e,f) The geometric mean and 95% confidence interval titre of (e) immunoglobulin (Ig)M or (f) IgG keyhole limpet haemocyanin (KLH)‐specific antibody after vaccination with keyhole limpet haemocyanin at baseline, weeks 4 and 8 started 12 weeks after ocrelizumab infusion (n = 34–68). (g–i) The response to: A/California/7/2009 (H1N1, n = 33–35); B/Phuket/3073/2013 (BPH, n = 31–33), A/Switzerland/9715293/2013 (H3N2, n = 27–30), B/Brisbane/60/2008 (BBR, n = 16–18), A/Hong Kong/4801/2014 (AHK, n = 5–6) influenza strain vaccination 12 weeks after ocrelizumab infusion was assessed. The results represent (g) the percentage of people with seroconversion, defined either a prevaccination haemagglutination inhibition (HI) titre < 10 and ≥ 40 at 4 weeks or a prevaccination ≥ 10 and at least a fourfold increase in HI titre, and seroprotection defined by titres > 40 at 4 weeks after vaccination. (h) The change in the geometric mean HI titres before and after vaccination (I) The percentage of people with a fourfold increase in strain‐specific > 40) at 4 weeks after vaccination.
Fig. 3
Fig. 3
Long‐term depletion of memory B cells induced by ocrelizumab. These data were extracted from the ocrelizumab Phase II clinical study report [105], supplied by the trial sponsor via the www.clinicaltrialdatarequest.com portal. (a) The data represents the mean percentage change from baseline, defined as the last observation up to the first day of ocrelizumab treatment. The subjects received placebo and three cycles of 600 mg ocrelizumab every 24 weeks. This was followed by a treatment‐free period to monitor B cell repletion in the Phase II extension study. The time represents the period from the last ocrelizumab infusion (n = 22–43). Naive [CD19+, CD21+, immunoglobulin (IgD)+, IgM+] and memory B cells (CD19+, CD27+, CD38low) and other immune subsets were assessed (n = 22–43/group). (b) Depletion of memory B cells was maintained during treatment. These data were obtained from people (n = 88) entering the open‐label extension (OLE) study after the treatment‐free period that had received three or four 6‐monthly cycles of at least 600 mg ocrelizumab to week 72, followed by a treatment‐free period to week 144, before entering the OLE phase where 600‐mg cycles of ocrelizumab were maintained at 24‐week intervals. The results represent the mean ± standard deviation of cells/μl (n = 22–69/group weeks 22–72). Although CD19 B cell numbers were consistent with the original levels, the baseline memory B cell levels failed to return to original levels at the start of the OLE. PMN = polymorphonuclear neutrophil.

References

    1. Du FH, Mills EA, Mao‐Draayer Y. Next‐generation anti‐CD20 monoclonal antibodies in autoimmune disease treatment. Auto Immun Highlights 2017; 8:12.
    1. Baker D, Marta M, Pryce G, Giovannoni G, Schmierer K. Memory B‐cells are major targets for effective immunotherapy in relapsing multiple sclerosis. EBioMedicine 2017; 16:41–50.
    1. Baker D, Pryce G, Amor S, Giovannoni G, Schmierer K. Learning from other autoimmunities to understand targeting of B‐cells to control multiple sclerosis. Brain 2018; 141:2824–8.
    1. Cang S, Mukhi N, Wang K, Liu D. Novel CD20 monoclonal antibodies for lymphoma therapy. J Hematol Oncol 2012; 5:64.
    1. Sabatino JJ, Zamvil SS, Hauser SL. B‐cell therapies in multiple sclerosis. Cold Spring Harb Perspect Med 2019; 9:a032037.
    1. Hauser SL, Bar‐Or A, Cohen J et al Efficacy and safety of ofatumumab versus teriflunomide in relapsing multiple sclerosis: results of the phase 3 ASCLEPIOS I and II trials. Mult Scler 2019; 25(Suppl 2):890–1.
    1. Fox E, Lovett‐Racke AE, Gormley M et al A phase 2 multicenter study of ublituximab, a novel glycoengineered anti‐CD20 monoclonal antibody, in patients with relapsing forms of multiple sclerosis. Mult Scler 2020. 10.1177/1352458520918375.
    1. Ineichen BV, Moridi T, Granberg T, Piehl F. Rituximab treatment for multiple sclerosis. Mult Scler 2020; 26:137–52.
    1. Baker D, Pryce G, James LK, Marta M, Schmierer K. The ocrelizumab phase II extension trial suggests the potential to improve the risk:benefit balance in multiple sclerosis. Mult Scler Relat Disord 2020. 10.1101/2020.01.09.20016774.
    1. Juto A, Fink K, Nimer F, Piehl F. Interrupting rituximab treatment in relapsing–remitting multiple sclerosis; no evidence of rebound disease activity. Mult Scler Relat Disord 2020; 37:101468.
    1. Trouvin AP, Jacquot S, Grigioni S et al Usefulness of monitoring of B‐cell depletion in rituximab‐treated rheumatoid arthritis patients in order to predict clinical relapse: a prospective observational study. Clin Exp Immunol 2015; 180:11–8.
    1. Kim SH, Kim W, Li XF, Jung IJ, Kim HJ. Repeated treatment with rituximab based on the assessment of peripheral circulating memory B‐cells in patients with relapsing neuromyelitis optica over 2 years. Arch Neurol 2011; 68:1412–20.
    1. Novi G, Fabbri S, Bovis F et al Tailoring B‐cells depleting therapy in MS according to memory B‐cells monitoring: a pilot study. P971. Mult Scler 2019; 25(Suppl 2):509–10.
    1. Marcinnò A, Marnetto F, Valentino P et al Rituximab‐induced hypogammaglobulinemia in patients with neuromyelitis optica spectrum disorders. Neurol Neuroimmunol Neuroinflamm 2018; 5:e498.
    1. Derfuss T, Weber MS, Hughes R et al Serum immunoglobulin levels and risk of serious infections in the pivotal Phase III trials of ocrelizumab in multiple sclerosis and their open‐label extensions. 65. Mult Scler 2019; 25(Suppl 2):20–1.
    1. Luna G, Alping P, Burman J et al Infection risks among patients with multiple sclerosis treated with fingolimod, natalizumab, rituximab, and injectable therapies. JAMA Neurol 2019; 77:184.
    1. Baloch S, Baloch MA, Zheng T, Pei X. The coronavirus disease 2019 (COVID‐19) pandemic. Tohoku J Exp Med 2020; 250:271–8.
    1. Huang C, Wang Y, Li X et al Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China [published correction appears in Lancet 2020 30 January]. Lancet 2020; 395:497–506.
    1. Brownlee W, Bourdette D, Broadley S, Killestein J, Ciccarelli O. Treating multiple sclerosis and neuromyelitis optica spectrum disorder during the COVID‐19 pandemic. Neurology 2020; 94:949–52.
    1. Baker D, Amor S, Kang AS, Schmierer K, Giovannoni G. The underpinning biology relating to multiple sclerosis disease modifying treatments during the COVID‐19 pandemic. Mult Scler Relat Disord 2020; 43:102174.
    1. Xu X, Chang XN, Pan HX et al Pathological changes of the spleen in ten patients with coronavirus disease 2019 (COVID‐19) by postmortem needle autopsy. Zhonghua Bing Li Xue Za Zhi 2020; 49:576–82.
    1. Henry BM, Vikse J, Benoit S, Favaloro EJ, Lippi G. Hyperinflammation and derangement of renin‐angiotensin‐aldosterone system in COVID‐19: A novel hypothesis for clinically suspected hypercoagulopathy and microvascular immunothrombosis. Clin Chim Acta 2020; 507:167–73.
    1. AminJafari A, Ghasemi S. The possible of immunotherapy for COVID‐19: a systematic review. Int Immunopharmacol 2020; 83:106455.
    1. Le Thanh T, Andreadakis Z, Kumar A et al The COVID‐19 vaccine development landscape. Nat Rev Drug Discov 2020; 19:305–6.
    1. Randolph HE, Barreiro LB. Herd immunity: understanding COVID‐19. Immunity 2020; 52:737–41.
    1. Bar‐Or A, Calabresi PA, Arnold D et al Rituximab in relapsing–remitting multiple sclerosis: a 72‐week, open‐label, phase I trial. Ann Neurol 2008;63:395–400.
    1. Channappanavar R, Fett C, Zhao J, Meyerholz DK, Perlman S. Virus‐specific memory CD8 T‐cells provide substantial protection from lethal severe acute respiratory syndrome coronavirus infection. J Virol 2014; 88:11034–44.
    1. Bao L, Deng W, Gao H et al Reinfection could not occur in SARS‐CoV‐2 infected rhesus macaques. bioRxiv 2020. 10.1101/2020.03.13.990226.
    1. Bao L, Deng W, Huang B et al The pathogenicity of SARS‐CoV‐2 in hACE2 transgenic mice. Nature 2020. 10.1038/s41586-020-2312-y.
    1. Giovannoni G, Hawkes C, Lechner‐Scott J, Levy M, Waubant E, Gold J. The COVID‐19 pandemic and the use of MS disease‐modifying therapies. Mult Scler Relat Disord 2020; 39:102073.
    1. Berger JR, Brandstadter R, Bar‐Or A. COVID‐19 and MS disease‐modifying therapies. Neurol Neuroimmunol Neuroinflamm 2020; 7:e761.
    1. Favalli EG, Ingegnoli F, De Lucia O, Cincinelli G, Cimaz R, Caporali R. COVID‐19 infection and rheumatoid arthritis: faraway, so close! Autoimmun Rev 2020; 19:102523.
    1. Wang B, Wang L, Kong X et al Long term coexistence of SARS‐CoV‐2 with antibody response in COVID‐19 patients. J Med Virol 2020. 10.1002/jmv.25946.
    1. Anand P, Slama MCC, Kaku M et al COVID‐19 in patients with myasthenia gravis. Muscle Nerve 2020. 10.1002/mus.26918.
    1. Dworakowska D, Grossman AB. Thyroid disease in the time of COVID‐19. Endocrine 2020; 68:471–4. 10.1007/s12020-020-02364-8.
    1. Salvarani C, Bajocchi G, Mancuso P et al Susceptibility and severity of COVID‐19 in patients treated with bDMARDS and tsDMARDs: a population‐based study. Ann Rheum Dis 2020. 10.1136/annrheumdis-2020-217903.
    1. Novi G, Mikulska M, Briano F et al COVID‐19 in a MS patient treated with ocrelizumab: does immunosuppression have a protective role? Mult Scler Relat Disord 2020; 15:102120.
    1. Louapre C, Maillart E, Roux T et al Patients with MS treated with immunosuppressive agents: across the COVID‐19 spectrum. Rev Neurol 2020; 176:523–5.
    1. Montero‐Escribano P, Matías‐Guiu J, Gómez‐Iglesias P, Porta‐Etessam J, Pytel V, Matias‐Guiu JA. Anti‐CD20 and COVID‐19 in multiple sclerosis and related disorders: a case series of 60 patients from Madrid, Spain. Mult Scler Relat Disord 2020; 42:102185.
    1. Safavi F, Nourbakhsh B, Azimi AR. B‐cell depleting therapies may affect susceptibility to acute respiratory illness among patients with multiple sclerosis during the early COVID‐19 epidemic in Iran. Mult Scler Relat Disord 2020; 43:102195.
    1. Hughes R, Pedotti R, Koendgen H. COVID‐19 in persons with multiple sclerosis treated with ocrelizumab – a pharmacovigilance case series. Mult Scler Relat Disord 2020; 42:102192.
    1. Suwanwongse K, Shabarek N. Benign course of COVID‐19 in a multiple sclerosis patient treated with ocrelizumab. Mult Scler Relat Disord 2020; 42:102201.
    1. Ghajarzadeh M, Mirmosayyeb O, Barzegar M et al Favorable outcome after COVID‐19 infection in a multiple sclerosis patient initiated on ocrelizumab during the pandemic. Mult Scler Relat Disord 2020; 43:102222.
    1. Chaudhry F, Bulka H, Rathnam AS et al COVID‐19 in multiple sclerosis patients and risk factors for severe infection. medRxiv 2020. 10.1101/2020.05.27.20114827.
    1. Sormani MP; Italian Study Group on COVID‐19 Infection in Multiple Sclerosis . An Italian programme for COVID‐19 infection in multiple sclerosis. Lancet Neurol 2020; 19:481–2.
    1. Salter A. iWiMS MS Covid‐19. 13 May 2020 ().
    1. Assmuth Oreja CS.iWiMS MS Covid‐19. 6 May 2020. Available at: .
    1. van der Welt A, Health A.iWiMS MS Covid‐19. 6 May 2020. Available at: .
    1. Louapre C, Collongues N, Stankoff B et al Clinical characteristics and outcomes in patients with coronavirus disease 2019 and multiple sclerosis. JAMA Neurol 2020. 10.1001/jamaneurol.2020.2581.
    1. Meca‐Lallana V, Aguirre C, del Río B, Cardeñoso L, Alarcon T, Vivancos J. COVID‐19 in 7 multiple sclerosis patients in treatment with anti CD20 therapies. Mult Scler Relat Disord 2020; 44:102306. accessed 20 May 2020.
    1. Conte WL. Attenuation of antibody response to SARS‐CoV‐2 in a patient on ocrelizumab with hypogammaglobulinemia. Mult Scler Relat Disord 2020; 44:102315.
    1. Rempe Thornton J, Harel A. Negative SARS‐CoV‐2 antibody testing following COVID‐19 infection in two MS patients treated with ocrelizumab. Mult Scler Rel Disord 2020. 10.1016/j.msard.2020.102341.
    1. Lucchini M, Bianco A, Del Giacomo P, De Fino C, Nociti V, Mirabella M. Is serological response to SARS‐CoV‐2 preserved in MS patients on ocrelizumab treatment? A case report. Mult Scler Relat Disord 2020. 10.1016/j.msard.2020.102323.
    1. Barzegar M, Mirmosayyeb O, Ghajarzadeh M et al Characteristics of COVID‐19 disease in multiple sclerosis patients. Mult Scler Relat Disord 2020; 45:102276.
    1. Hillert J. iWiMS MS Covid‐19. 20 May 2020. Available at: .
    1. Parrotta E, Kister I, Charvet L et al COVID‐19 outcomes in MS: early experience from NYU multiple sclerosis comprehensive care center. medRxiv 2020. 10.1101/2020.05.12.20094508.
    1. Suthar MS, Zimmerman M, Kauffman R et al Rapid generation of neutralizing antibody responses in COVID‐19 patients. medRxiv 2020. 10.1101/2020.05.03.20084442.
    1. Bertoglio F, Meier D, Langreder N et al SARS‐CoV‐2 neutralizing human recombinant antibodies selected from pre‐pandemic healthy donors binding at RBD–ACE2 interface. bioRxiv 2020. 10.1101/2020.06.05.135921.
    1. Zeng QL, Yu ZJ, Gou JJ et al Effect of convalescent plasma therapy on viral shedding and survival in COVID‐19 patients. J Infect Dis 2020:jiaa228.
    1. Ye M, Fu D, Ren Y et al Treatment with convalescent plasma for COVID‐19 patients in Wuhan, China. J Med Virol 2020. 10.1002/jmv.25882.
    1. Soresina A, Moratto D, Chiarini M et al Two X‐linked agammaglobulinemia patients develop pneumonia as COVID‐19 manifestation but recover. Pediatr Allergy Immunol 2020. 10.1111/pai.13263.
    1. Quinti I, Lougaris V, Milito C et al A possible role for B‐cells in COVID‐19? Lesson from patients with agammaglobulinemia. J Allergy Clin Immunol 2020; 146:211–3.e4.
    1. Ackermann M, Verleden SE, Kuehnel M et al Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in Covid‐19. N Engl J Med 2020. 10.1056/NEJMoa2015432.
    1. Tseng YH, Yang RC, Lu TS. Two hits to the renin–angiotensin system may play a key role in severe COVID‐19. Kaohsiung J Med Sci 2020. 10.1002/kjm2.12237.
    1. Monti S, Balduzzi S, Delvino P, Bellis E, Quadrelli VERSUS, Montecucco C. Clinical course of COVID‐19 in a series of patients with chronic arthritis treated with immunosuppressive targeted therapies. Ann Rheum Dis 2020; 79:667–8.
    1. Gianfrancesco M, Hyrich KL, Al‐Adely S et al Characteristics associated with hospitalisation for COVID‐19 in people with rheumatic disease: data from the COVID‐19 Global Rheumatology Alliance physician‐reported registry. Ann Rheum Dis 2020. 10.1136/annrheumdis-2020-217871.
    1. Ocrevus ® European public assessment report . 7/01/2019 update. Summary of product characteristics. Available at: accessed 5 January 2020.
    1. Fallet B, Kyburz D, Walker UA. Mild course of coronavirus disease 2019 and spontaneous severe acute respiratory syndrome coronavirus 2 clearance in a patient with depleted peripheral blood B‐cells due to treatment with rituximab. Arthritis Rheum 2020; 2020 10.1002/art.41380.
    1. Xiang F, Wang X, He X et al Antibody detection and dynamic characteristics in patients with COVID‐19. Clin Infect Dis 2020. 10.1093/cid/ciaa461.
    1. Shen L, Wang C, Zhao J et al Delayed specific IgM antibody responses observed among COVID‐19 patients with severe progression. Emerg Microbes Infect 2020; 9:1096–101.
    1. Yu HQ, Sun BQ, Fang ZF et al Distinct features of SARS‐CoV‐2‐specific IgA response in COVID‐19 patients. Eur Respir J 2020; 8:2001526.
    1. Dahlke C, Heidepriem J, Kobbe R et al Distinct early IgA profile may determine severity of COVID‐19 symptoms: an immunological case series. medRxiv 2020. 10.1101/2020.04.14.20059733.
    1. Grifoni A, Weiskopf D, Ramirez SI et al Targets of T cell responses to SARS‐CoV‐2 coronavirus in humans with COVID‐19 disease and unexposed individuals. Cell 2020; 181:1489–1501.e15.
    1. Ng K, Faulkner N, Cornish G et al Pre‐existing and de novo humoral immunity to SARS‐CoV‐2 in humans. bioRxiv 2020. 10.1101/2020.05.14.095414.
    1. Che XY, Qiu LW, Liao ZY et al Antigenic cross‐reactivity between severe acute respiratory syndrome‐associated coronavirus and human coronaviruses 229E and OC43. J Infect Dis 2005; 191:2033–7.
    1. Negro F. Is antibody‐dependent enhancement playing a role in COVID‐19 pathogenesis? Swiss Med Wkly 2020; 150:w20249.
    1. Magro C, Mulvey JJ, Berlin D et al Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID‐19 infection: a report of five cases. Transl Res 2020; 5244:1–13.
    1. Xiao T, Wang Y, Yuan J et al Early viral clearance and antibody kinetics of COVID‐19 among asymptomatic carriers. medRxiv 2020. 10.1101/2020.04.28.20083139.
    1. Brochot E, Demey B, Touze A et al Anti‐spike, anti‐nucleocapsid and neutralizing antibodies in SARS‐CoV‐2 inpatients and asymptomatic carriers. medRxiv 2020. 10.1101/2020.05.12.20098236.
    1. Liu T, Wu S, Tao H et al. Prevalence of IgG antibodies to SARS‐CoV‐2 in Wuhan – implications for the ability to produce long‐lasting protective antibodies against SARS‐CoV‐2. medRxiv 2020. 10.1101/2020.06.13.20130252.
    1. Galanti M, Shaman J. Seasonal cold‐inducing coronavirus can be repeatedly detected in some individuals. medRxiv 2020. 10.1101/2020.04.27.20082032.
    1. Ravioli S, Ochsner H, Lindner G. Reactivation of COVID‐19 pneumonia: a report of two cases. J Infect 2020. 10.1016/j.jinf.2020.05.008.
    1. Wang QX, Huang KC, Qi L, Zeng XH, Zheng SL. No infectious risk of COVID‐19 patients with long‐term fecal 2019‐nCoV nucleic acid positive. Eur Rev Med Pharmacol Sci 2020; 24:5772–7.
    1. Korean Centre for Disease Control and Prevention . Findings from investigation and analysis of re‐positive cases. Notice 2020–05‐19~2020‐12‐31. accessed 19 May 2020. Available at: .
    1. Chandrashekar A, Liu J, Martinot AJ et al SARS‐CoV‐2 infection protects against rechallenge in rhesus macaques. Science 2020. 10.1126/science.abc4776.
    1. Yu J, Tostanoski LH, Peter L et al DNA vaccine protection against SARS‐CoV‐2 in rhesus macaques. Science 2020. 10.1126/science.abc6284.
    1. Zhu FC, Li YH, Guan XH et al Safety, tolerability, and immunogenicity of a recombinant adenovirus type‐5 vectored COVID‐19 vaccine: a dose‐escalation, open‐label, non‐randomised, first‐in‐human trial. Lancet 2020; 395:1845–54.
    1. Wu LP, Wang NC, Chang YH et al Duration of antibody responses after severe acute respiratory syndrome. Emerg Infect Dis 2007; 13:1562–4.
    1. Cioc AM, Vanderwerf SM, Peterson BA, Robu VG, Forster CL, Pambuccian SE. Rituximab‐induced changes in hematolymphoid tissues found at autopsy. Am J Clin Pathol 2008; 130:604–12.
    1. Ramwadhdoebe TH, van Baarsen LGM, Boumans MJH et al Effect of rituximab treatment on T and B‐cell subsets in lymph node biopsies of patients with rheumatoid arthritis. Rheumatology 2019; 58:1075–85.
    1. Stokmaier D, Winthrop K, Chognot C et al Effect of ocrelizumab on vaccine responses in patients with multiple sclerosis (S36.002). Neurology 2018; 90(15 Suppl):S36.002.
    1. Simonsen O, Bentzon MW, Heron I. ELISA for the routine determination of antitoxic immunity to tetanus. J Biol Standard 1986; 14:231–9.
    1. Kim W, Kim SH, Huh SY et al Reduced antibody formation after influenza vaccination in patients with neuromyelitis optica spectrum disorder treated with rituximab. Eur J Neurol 2013;20:975–80.
    1. Bingham CO, Looney RJ, Deodhar A et al Immunization responses in rheumatoid arthritis patients treated with rituximab: results from a controlled clinical trial. Arthritis Rheum 2010; 62:64–74.
    1. Nazi I, Kelton JG, Larché M et al The effect of rituximab on vaccine responses in patients with immune thrombocytopenia. Blood 2013; 122:1946–53.
    1. van Assen S, Holvast A, Telgt DS et al Patients with humoral primary immunodeficiency do not develop protective anti‐influenza antibody titers after vaccination with trivalent subunit influenza vaccine. Clin Immunol 2010; 136:228–35.
    1. Richi P, Alonso O, Martín MD et al Evaluation of the immune response to hepatitis B vaccine in patients on biological therapy: results of the RIER cohort study. Clin Rheumatol 2020. 10.1007/s10067-020-05042-2.
    1. Cho A, Bradley B, Kauffman R et al Robust memory responses against influenza vaccination in pemphigus patients previously treated with rituximab. JCI Insight 2017; 2:e93222.
    1. Palanichamy A, Jahn S, Nickles D et al Rituximab efficiently depletes increased CD20‐expressing T‐cells in multiple sclerosis patients. J Immunol 2014; 193:580–6.
    1. Baker D, Herrod SS, Alvarez‐Gonzalez C, Giovannoni G, Schmierer K. Interpreting lymphocyte reconstitution data from the pivotal phase 3 trials of alemtuzumab. JAMA Neurol 2017; 74:961–9.
    1. Ziemssen T, Bar‐Or A, Arnold DL et al P 2 Effect of ocrelizumab on humoral immunity markers in the phase iii, double‐blind, double‐dummy, IFN β ‐1a–controlled OPERA I and OPERA II studies. Clin Neurophysiol 2017; 128:e326–e327.
    1. Roll P, Palanichamy A, Kneitz C, Dorner T, Tony HP. Regeneration of B‐cell subsets after transient B‐cell depletion using anti‐CD20 antibodies in rheumatoid arthritis. Arthritis Rheum 2006; 54:2377–86.
    1. Akgün K, Blankenburg J, Marggraf M, Haase R, Ziemssen T. Event‐driven immunoprofiling predicts return of disease activity in alemtuzumab‐treated multiple sclerosis. Front Immunol 2020; 11:56.
    1. Bar‐Or A, Grove RA, Austin DJ et al Subcutaneous ofatumumab in patients with relapsing‐remitting multiple sclerosis: the MIRROR study. Neurology 2018; 90:e1805–e1814.
    1. WA21493 Clinical study report 2016 . WA21493‐Phase II, multicenter, randomized parallel‐group, partially blinded, placebo, Avonex® controlled dose finding study to evaluate the efficacy as measured by brain MRI lesions and safety of 2 dose regimens of ocrelizumab in patients with RRMS. Report no. 1062910. March 2016.
    1. Signoriello E, Bonavita S, Di Pietro A et al BMI influences CD20 kinetics in multiple sclerosis patients treated with ocrelizumab. Mult Scler Relat Disord 2020; 43:102186.
    1. Kletzl H, Gibiansky E, Petry C et al Pharmacokinetics, pharmacodynamics and exposure‐response analyses of ocrelizumab in patients with multiple sclerosis. Neurol 2019; 92(Suppl 15):N4.001.
    1. Comi G, Cook S, Giovannoni G et al Effect of cladribine tablets on lymphocyte reduction and repopulation dynamics in patients with relapsing multiple sclerosis. Mult Scler Relat Disord 2019; 29:168–74.
    1. Hermann R, Karlsson MO, Novakovic AM, Terranova N, Fluck M, Munafo A. The clinical pharmacology of cladribine tablets for the treatment of relapsing multiple sclerosis. Clin Pharmokinet 2019; 58:283–97.
    1. Baker D, Pryce G, Herrod SS, Schmierer K. Potential mechanisms of action related to the efficacy and safety of cladribine. Mult Scler Relat Disord 2019; 30:176–86.
    1. Li Z, Richards S, Surks HK, Jacobs A, Panzara MA. Clinical pharmacology of alemtuzumab, an anti‐CD52 immunomodulator, in multiple sclerosis. Clin Exp Immunol 2018; 194:295–314.
    1. McCarthy CL, Tuohy O, Compston DA, Kumararatne DS, Coles AJ, Jones JL. Immune competence after alemtuzumab treatment of multiple sclerosis. Neurology 2013; 81:872–6.
    1. Gingele S, Jacobus TL, Konen FF et al Ocrelizumab depletes CD20⁺ T cells in multiple sclerosis patients. Cells 2018; 8:12.
    1. Parrino J, McNeil SA, Lawrence SJ et al Safety and immunogenicity of inactivated varicella‐zoster virus vaccine in adults with hematologic malignancies receiving treatment with anti‐CD20 monoclonal antibodies. Vaccine 2017; 35:1764–9.
    1. Ng OW, Chia A, Tan AT et al Memory T‐cell responses targeting the SARS coronavirus persist up to 11 years post‐infection. Vaccine 2016; 34:2008–14.
    1. Robbiani DF, Gaebler C, Muecksch F et al Convergent antibody responses to SARS‐CoV‐2 in convalescent individuals. Nature 2020. 10.1038/s41586-020-2456-9.
    1. Gallais F, Velay A, Wendling MJ, Nazon C, Partisani M, Sibilia J, Candon S, Fafi‐Kremer S. Intrafamilial exposure to SARS‐CoV‐2 induces cellular immune response without seroconversion. medRxiv 2020. 10.1101/2020.06.21.20132449.
    1. Sekine T, Perez‐Potti A, Rivera‐Ballesteros O, et al Robust T cell immunity in convalescent individuals with asymptomatic or mild COVID‐19. bioRxiv 2020. 10.1101/2020.06.29.174888.

Source: PubMed

3
Abonnere