B Cell Kinetics upon Therapy Commencement for Active Extrarenal Systemic Lupus Erythematosus in Relation to Development of Renal Flares: Results from Three Phase III Clinical Trials of Belimumab

Ioannis Parodis, Alvaro Gomez, Julius Lindblom, Jun Weng Chow, Christopher Sjöwall, Savino Sciascia, Mariele Gatto, Ioannis Parodis, Alvaro Gomez, Julius Lindblom, Jun Weng Chow, Christopher Sjöwall, Savino Sciascia, Mariele Gatto

Abstract

Renal flares constitute major determinants of poor prognosis in people living with systemic lupus erythematosus (SLE). The aim of the present study was to investigate changes in B cell subsets in relation to renal flares upon initiation of standard therapy (ST) plus belimumab or placebo in patients with SLE. Using data from the BLISS-76, BLISS-SC, and BLISS Northeast Asia trials, we investigated associations of relative to baseline rapid (through week 8) and early (through week 24) percentage changes in circulating CD19+ B cell subsets characterised through flow cytometry, anti-dsDNA antibodies, and complement levels with the occurrence of renal flares over one year. Patients who developed renal flares showed more prominent rapid decreases in CD19+CD20+CD138+ short-lived plasma cells (-50.4% vs. -16.7%; p = 0.019) and CD19+CD20-CD27bright plasmablasts (-50.0% vs. -29.9%; p = 0.020) compared to non-flaring patients, followed by a subsequent return. Less prominent rapid reductions in CD19+CD27-CD24brightCD38bright transitional B cells (-42.9% vs. -75.0%; p = 0.038) and CD19+CD20-CD138+ peripheral long-lived plasma cells (-11.3% vs. -29.2%; p = 0.019) were seen in belimumab-treated-but not placebo-treated-patients who developed renal flares compared to belimumab-treated patients who did not. Rapid and early changes in anti-dsDNA or complement levels showed no clear association with renal flares. In summary, a rapid drop followed by a subsequent return in circulating short-lived plasma cells and plasmablasts upon treatment for active extra-renal SLE portended renal flares, indicating a need for therapeutic adjustments in patients showing such B cell patterns. Rapid decreases in transitional B cells and peripheral long-lived plasma cells upon belimumab therapy commencement may signify a greater protection against renal flares. B cell kinetics may prove useful in early drug evaluation.

Keywords: B cells; B lymphocyte; belimumab; biologics; biomarkers; plasma cells; renal flares; systemic lupus erythematosus.

Conflict of interest statement

IP has received research funding and/or honoraria from Amgen, AstraZeneca, Aurinia Pharmaceuticals, Elli Lilly and Company, Gilead Sciences, GlaxoSmithKline, Janssen Pharmaceuticals, Novartis, and F. Hoffmann-La Roche AG. The other authors declare that they have no conflicts of interest related to this work. The funders had no role in the design of the study, the analyses or interpretation of data, or the writing of the manuscript.

Figures

Figure 1
Figure 1
B cell alterations in relation to renal flares. The graphs delineate relative to baseline percentage changes in selected B cell and peripheral plasma cell subsets in patients who developed at least one renal flare during the study period (continuous lines) and patients who did not (dashed lines). Comparisons between patients who flared and patients who did not were conducted using multivariable logistic regression analysis to account for potential confounders and are illustrated for patients who received non-biological standard therapy plus belimumab (red lines) and patients who received non-biological standard therapy alone (blue lines). Comparisons between treatment arms were conducted using non-parametrical Mann–Whitney U tests. Whiskers indicate the interquartile range of distributions. The number of patients with available data at each time point is indicated for each patient subgroup.
Figure 2
Figure 2
Transitional and naïve B cell alterations in relation to renal flares. The graphs delineate relative to baseline percentage changes in transitional and naïve B cell subsets in patients who developed at least one renal flare during the study period (continuous lines) and patients who did not (dashed lines) in a subanalysis of data from the BLISS-SC trial. Comparisons between patients who flared and patients who did not were conducted for patients with available data, stratified into patients who received non-biological standard therapy plus belimumab (red lines) and patients who received non-biological standard therapy alone (blue lines). P values are derived from non-parametric Mann–Whitney U tests. Whiskers indicate the interquartile range of distributions. The number of patients with available data at each time point is indicated for each patient subgroup.
Figure 3
Figure 3
Changes in serological markers in relation to renal flares. The graphs delineate relative to baseline percentage changes in anti-dsDNA, C3, and C4 levels in patients who developed at least one renal flare during the study period (continuous lines) and patients who did not (dashed lines). Comparisons between patients who flared and patients who did not were conducted using multivariable logistic regression analysis to account for potential confounders and are illustrated for patients who received non-biological standard therapy plus belimumab (red lines) and patients who received non-biological standard therapy alone (blue lines). For anti-dsDNA levels, a separate analysis for patients with positive anti-dsDNA levels (≥30 IU/mL) at baseline is also demonstrated. Comparisons between treatment arms were conducted using non-parametrical Mann–Whitney U tests. Whiskers indicate the interquartile range of distributions. The number of patients with available data at each time point is indicated for each patient subgroup. Anti-dsDNA: anti-double-stranded DNA antibodies; C3: complement component 3; C4: complement component 4.

References

    1. Gasparotto M., Gatto M., Binda V., Doria A., Moroni G. Lupus nephritis: Clinical presentations and outcomes in the 21st century. Rheumatology (Oxford) 2020;59:v39–v51. doi: 10.1093/rheumatology/keaa381.
    1. Moroni G., Vercelloni P.G., Quaglini S., Gatto M., Gianfreda D., Sacchi L., Raffiotta F., Zen M., Costantini G., Urban M.L., et al. Changing patterns in clinical-histological presentation and renal outcome over the last five decades in a cohort of 499 patients with lupus nephritis. Ann. Rheum. Dis. 2018;77:1318–1325. doi: 10.1136/annrheumdis-2017-212732.
    1. Ugarte-Gil M.F., Acevedo-Vásquez E., Alarcón G.S., Pastor-Asurza C.A., Alfaro-Lozano J.L., Cucho-Venegas J.M., Segami M.I., Wojdyla D., Soriano E.R., Drenkard C., et al. The number of flares patients experience impacts on damage accrual in systemic lupus erythematosus: Data from a multiethnic Latin American cohort. Ann. Rheum. Dis. 2015;74:1019–1023. doi: 10.1136/annrheumdis-2013-204620.
    1. Doria A., Amoura Z., Cervera R., Khamastha M.A., Schneider M., Richter J., Guillemin F., Kobelt G., Maurel F., Garofano A., et al. Annual direct medical cost of active systemic lupus erythematosus in five European countries. Ann. Rheum. Dis. 2014;73:154–160. doi: 10.1136/annrheumdis-2012-202443.
    1. Anders H.J., Saxena R., Zhao M.H., Parodis I., Salmon J.E., Mohan C. Lupus nephritis. Nat. Rev. Dis. Prim. 2020;6:7. doi: 10.1038/s41572-019-0141-9.
    1. Isenberg D.A., Allen E., Farewell V., D’Cruz D., Alarcon G.S., Aranow C., Bruce I.N., Dooley M.A., Fortin P.R., Ginzler E.M., et al. An assessment of disease flare in patients with systemic lupus erythematosus: A comparison of BILAG 2004 and the flare version of SELENA. Ann. Rheum. Dis. 2011;70:54–59. doi: 10.1136/ard.2010.132068.
    1. Ligtenberg G., Arends S., Stegeman C.A., de Leeuw K. Predictors of renal flares and long-term renal outcome in patients with lupus nephritis: Results from daily clinical practice. Clin. Exp. Rheumatol. 2021;40:33–38. doi: 10.55563/clinexprheumatol/c58c39.
    1. Luís M.S.F., Bultink I.E.M., da Silva J.A.P., Voskuyl A.E., Inês L.S. Early predictors of renal outcome in patients with proliferative lupus nephritis: A 36-month cohort study. Rheumatology (Oxford) 2021;60:5134–5141. doi: 10.1093/rheumatology/keab126.
    1. Pisetsky D.S., Lipsky P.E. New insights into the role of antinuclear antibodies in systemic lupus erythematosus. Nat. Rev. Rheumatol. 2020;16:565–579. doi: 10.1038/s41584-020-0480-7.
    1. Gensous N., Marti A., Barnetche T., Blanco P., Lazaro E., Seneschal J., Truchetet M.E., Duffau P., Richez C. Predictive biological markers of systemic lupus erythematosus flares: A systematic literature review. Arthritis. Res. Ther. 2017;19:238. doi: 10.1186/s13075-017-1442-6.
    1. Enocsson H., Sjöwall C., Wirestam L., Dahle C., Kastbom A., Rönnelid J., Wetterö J., Skogh T. Four Anti-dsDNA Antibody Assays in Relation to Systemic Lupus Erythematosus Disease Specificity and Activity. J. Rheumatol. 2015;42:817–825. doi: 10.3899/jrheum.140677.
    1. Parodis I., Stockfelt M., Sjowall C. B Cell Therapy in Systemic Lupus Erythematosus: From Rationale to Clinical Practice. Front. Med. 2020;7:316. doi: 10.3389/fmed.2020.00316.
    1. Gatto M., Saccon F., Andreoli L., Bartoloni E., Benvenuti F., Bortoluzzi A., Bozzolo E., Brunetta E., Canti V., Cardinaletti P., et al. Durable renal response and safety with add-on belimumab in patients with lupus nephritis in real-life setting (BeRLiSS-LN). Results from a large, nationwide, multicentric cohort. J. Autoimmun. 2021;124:102729. doi: 10.1016/j.jaut.2021.102729.
    1. Gatto M., Saccon F., Zen M., Regola F., Fredi M., Andreoli L., Tincani A., Urban M.L., Emmi G., Ceccarelli F., et al. Early Disease and Low Baseline Damage as Predictors of Response to Belimumab in Patients WITH Systemic Lupus Erythematosus in a Real-Life Setting. Arthritis Rheumatol. 2020;72:1314–1324. doi: 10.1002/art.41253.
    1. Sciascia S., Radin M., Yazdany J., Levy R.A., Roccatello D., Dall’Era M., Cuadrado M.J. Efficacy of belimumab on renal outcomes in patients with systemic lupus erythematosus: A systematic review. Autoimmun. Rev. 2017;16:287–293. doi: 10.1016/j.autrev.2017.01.010.
    1. Iaccarino L., Bettio S., Reggia R., Zen M., Frassi M., Andreoli L., Gatto M., Piantoni S., Nalotto L., Franceschini F., et al. Effects of Belimumab on Flare Rate and Expected Damage Progression in Patients With Active Systemic Lupus Erythematosus. Arthritis Care Res. (Hoboken) 2017;69:115–123. doi: 10.1002/acr.22971.
    1. Parodis I., Sjowall C., Jonsen A., Ramskold D., Zickert A., Frodlund M., Sohrabian A., Arnaud L., Ronnelid J., Malmstrom V., et al. Smoking and pre-existing organ damage reduce the efficacy of belimumab in systemic lupus erythematosus. Autoimmun. Rev. 2017;16:343–351. doi: 10.1016/j.autrev.2017.02.005.
    1. Dooley M.A., Houssiau F., Aranow C., D’Cruz D.P., Askanase A., Roth D.A., Zhong Z.J., Cooper S., Freimuth W.W., Ginzler E.M., et al. Effect of belimumab treatment on renal outcomes: Results from the phase 3 belimumab clinical trials in patients with SLE. Lupus. 2013;22:63–72. doi: 10.1177/0961203312465781.
    1. Furie R., Rovin B.H., Houssiau F., Malvar A., Teng Y.K.O., Contreras G., Amoura Z., Yu X., Mok C.C., Santiago M.B., et al. Two-Year, Randomized, Controlled Trial of Belimumab in Lupus Nephritis. N. Engl. J. Med. 2020;383:1117–1128. doi: 10.1056/NEJMoa2001180.
    1. Parodis I., Houssiau F.A. From sequential to combination and personalised therapy in lupus nephritis: Moving towards a paradigm shift? Ann. Rheum. Dis. 2021;81:15–19. doi: 10.1136/annrheumdis-2021-221270.
    1. Parodis I., Vital E.M., Hassan S.U., Jonsen A., Bengtsson A.A., Eriksson P., Leonard D., Gunnarsson I., Ronnblom L., Sjowall C. De novo lupus nephritis during treatment with belimumab. Rheumatology (Oxford) 2021;60:4348–4354. doi: 10.1093/rheumatology/keaa796.
    1. Jacobi A.M., Huang W., Wang T., Freimuth W., Sanz I., Furie R., Mackay M., Aranow C., Diamond B., Davidson A. Effect of long-term belimumab treatment on B cells in systemic lupus erythematosus: Extension of a phase II, double-blind, placebo-controlled, dose-ranging study. Arthritis Rheum. 2010;62:201–210. doi: 10.1002/art.27189.
    1. Stohl W., Hiepe F., Latinis K.M., Thomas M., Scheinberg M.A., Clarke A., Aranow C., Wellborne F.R., Abud-Mendoza C., Hough D.R., et al. Belimumab reduces autoantibodies, normalizes low complement levels, and reduces select B cell populations in patients with systemic lupus erythematosus. Arthritis Rheum. 2012;64:2328–2337. doi: 10.1002/art.34400.
    1. Ramskold D., Parodis I., Lakshmikanth T., Sippl N., Khademi M., Chen Y., Zickert A., Mikes J., Achour A., Amara K., et al. B cell alterations during BAFF inhibition with belimumab in SLE. EBioMedicine. 2019;40:517–527. doi: 10.1016/j.ebiom.2018.12.035.
    1. Arends E.J., Zlei M., Tipton C.M., Osmani Z., Kamerling S., Rabelink T., Sanz I., Van Dongen J.J.M., Van Kooten C., Teng Y.K.O. POS0680 Belimumab Add-On Therapy Mobilises Memory B Cells into the Circulation of Patients with SLE. Ann. Rheum. Dis. 2021;80:585. doi: 10.1136/annrheumdis-2021-eular.248.
    1. Parodis I., Tamirou F., Houssiau F.A. Prediction of prognosis and renal outcome in lupus nephritis. Lupus Sci. Med. 2020;7:e000389. doi: 10.1136/lupus-2020-000389.
    1. Dall’Era M., Cisternas M.G., Smilek D.E., Straub L., Houssiau F.A., Cervera R., Rovin B.H., Mackay M. Predictors of long-term renal outcome in lupus nephritis trials: Lessons learned from the Euro-Lupus Nephritis cohort. Arthritis Rheumatol. 2015;67:1305–1313. doi: 10.1002/art.39026.
    1. Moroni G., Quaglini S., Maccario M., Banfi G., Ponticelli C. “Nephritic flares” are predictors of bad long-term renal outcome in lupus nephritis. Kidney Int. 1996;50:2047–2053. doi: 10.1038/ki.1996.528.
    1. Tamirou F., Lauwerys B.R., Dall’Era M., Mackay M., Rovin B., Cervera R., Houssiau F.A., Investigators M.N.T. A proteinuria cut-off level of 0.7 g/day after 12 months of treatment best predicts long-term renal outcome in lupus nephritis: Data from the MAINTAIN Nephritis Trial. Lupus Sci. Med. 2015;2:e000123. doi: 10.1136/lupus-2015-000123.
    1. Ugolini-Lopes M.R., Seguro L.P.C., Castro M.X.F., Daffre D., Lopes A.C., Borba E.F., Bonfa E. Early proteinuria response: A valid real-life situation predictor of long-term lupus renal outcome in an ethnically diverse group with severe biopsy-proven nephritis? Lupus Sci. Med. 2017;4:e000213. doi: 10.1136/lupus-2017-000213.
    1. Parodis I., Adamichou C., Aydin S., Gomez A., Demoulin N., Weinmann-Menke J., Houssiau F.A., Tamirou F. Per-protocol repeat kidney biopsy portends relapse and long-term outcome in incident cases of proliferative lupus nephritis. Rheumatology (Oxford) 2020;59:3424–3434. doi: 10.1093/rheumatology/keaa129.
    1. Espeli M., Bokers S., Giannico G., Dickinson H.A., Bardsley V., Fogo A.B., Smith K.G. Local renal autoantibody production in lupus nephritis. J. Am. Soc. Nephrol. 2011;22:296–305. doi: 10.1681/ASN.2010050515.
    1. Gatto M., Radu C.M., Luisetto R., Ghirardello A., Bonsembiante F., Trez D., Valentino S., Bottazzi B., Simioni P., Cavicchioli L., et al. Immunization with Pentraxin3 prevents transition from subclinical to clinical lupus nephritis in lupus-prone mice: Insights from renal ultrastructural findings. J. Autoimmun. 2020;111:102443. doi: 10.1016/j.jaut.2020.102443.
    1. Sekine H., Watanabe H., Gilkeson G.S. Enrichment of anti-glomerular antigen antibody-producing cells in the kidneys of MRL/MpJ-Fas(lpr) mice. J. Immunol. 2004;172:3913–3921. doi: 10.4049/jimmunol.172.6.3913.
    1. Chang A., Henderson S.G., Brandt D., Liu N., Guttikonda R., Hsieh C., Kaverina N., Utset T.O., Meehan S.M., Quigg R.J., et al. In situ B cell-mediated immune responses and tubulointerstitial inflammation in human lupus nephritis. J. Immunol. 2011;186:1849–1860. doi: 10.4049/jimmunol.1001983.
    1. Hutloff A., Buchner K., Reiter K., Baelde H.J., Odendahl M., Jacobi A., Dorner T., Kroczek R.A. Involvement of inducible costimulator in the exaggerated memory B cell and plasma cell generation in systemic lupus erythematosus. Arthritis Rheum. 2004;50:3211–3220. doi: 10.1002/art.20519.
    1. Ramanujam M., Bethunaickan R., Huang W., Tao H., Madaio M.P., Davidson A. Selective blockade of BAFF for the prevention and treatment of systemic lupus erythematosus nephritis in NZM2410 mice. Arthritis Rheum. 2010;62:1457–1468. doi: 10.1002/art.27368.
    1. Bekar K.W., Owen T., Dunn R., Ichikawa T., Wang W., Wang R., Barnard J., Brady S., Nevarez S., Goldman B.I., et al. Prolonged effects of short-term anti-CD20 B cell depletion therapy in murine systemic lupus erythematosus. Arthritis Rheum. 2010;62:2443–2457. doi: 10.1002/art.27515.
    1. Jonsdottir T., Zickert A., Sundelin B., Henriksson E.W., van Vollenhoven R.F., Gunnarsson I. Long-term follow-up in lupus nephritis patients treated with rituximab--clinical and histopathological response. Rheumatology (Oxford) 2013;52:847–855. doi: 10.1093/rheumatology/kes348.
    1. Sfikakis P.P., Boletis J.N., Lionaki S., Vigklis V., Fragiadaki K.G., Iniotaki A., Moutsopoulos H.M. Remission of proliferative lupus nephritis following B cell depletion therapy is preceded by down-regulation of the T cell costimulatory molecule CD40 ligand: An open-label trial. Arthritis Rheum. 2005;52:501–513. doi: 10.1002/art.20858.
    1. Furie R.A., Aroca G., Cascino M.D., Garg J.P., Rovin B.H., Alvarez A., Fragoso-Loyo H., Zuta-Santillan E., Schindler T., Brunetta P., et al. B-cell depletion with obinutuzumab for the treatment of proliferative lupus nephritis: A randomised, double-blind, placebo-controlled trial. Ann. Rheum. Dis. 2022;81:100–107. doi: 10.1136/annrheumdis-2021-220920.
    1. Chan O.T., Hannum L.G., Haberman A.M., Madaio M.P., Shlomchik M.J. A novel mouse with B cells but lacking serum antibody reveals an antibody-independent role for B cells in murine lupus. J. Exp. Med. 1999;189:1639–1648. doi: 10.1084/jem.189.10.1639.
    1. Regola F., Piantoni S., Lowin T., Archetti S., Reggia R., Kumar R., Franceschini F., Airò P., Tincani A., Andreoli L., et al. Association Between Changes in BLyS Levels and the Composition of B and T Cell Compartments in Patients With Refractory Systemic Lupus Erythematosus Treated With Belimumab. Front. Pharmacol. 2019;10:433. doi: 10.3389/fphar.2019.00433.
    1. Huang W., Quach T.D., Dascalu C., Liu Z., Leung T., Byrne-Steele M., Pan W., Yang Q., Han J., Lesser M., et al. Belimumab promotes negative selection of activated autoreactive B cells in systemic lupus erythematosus patients. JCI Insight. 2018;3:e122525. doi: 10.1172/jci.insight.122525.
    1. Staveri C., Karokis D., Liossis S.C. New onset of lupus nephritis in two patients with SLE shortly after initiation of treatment with belimumab. Semin. Arthritis Rheum. 2017;46:788–790. doi: 10.1016/j.semarthrit.2016.09.006.
    1. Parodis I., Akerstrom E., Sjowall C., Sohrabian A., Jonsen A., Gomez A., Frodlund M., Zickert A., Bengtsson A.A., Ronnelid J., et al. Autoantibody and Cytokine Profiles during Treatment with Belimumab in Patients with Systemic Lupus Erythematosus. Int. J. Mol. Sci. 2020;21:3463. doi: 10.3390/ijms21103463.
    1. Furie R., Petri M., Zamani O., Cervera R., Wallace D.J., Tegzova D., Sanchez-Guerrero J., Schwarting A., Merrill J.T., Chatham W.W., et al. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011;63:3918–3930. doi: 10.1002/art.30613.
    1. Stohl W., Schwarting A., Okada M., Scheinberg M., Doria A., Hammer A.E., Kleoudis C., Groark J., Bass D., Fox N.L., et al. Efficacy and Safety of Subcutaneous Belimumab in Systemic Lupus Erythematosus: A Fifty-Two-Week Randomized, Double-Blind, Placebo-Controlled Study. Arthritis Rheumatol. 2017;69:1016–1027. doi: 10.1002/art.40049.
    1. Zhang F., Bae S.C., Bass D., Chu M., Egginton S., Gordon D., Roth D.A., Zheng J., Tanaka Y. A pivotal phase III, randomised, placebo-controlled study of belimumab in patients with systemic lupus erythematosus located in China, Japan and South Korea. Ann. Rheum. Dis. 2018;77:355–363. doi: 10.1136/annrheumdis-2017-211631.
    1. Petri M., Kim M.Y., Kalunian K.C., Grossman J., Hahn B.H., Sammaritano L.R., Lockshin M., Merrill J.T., Belmont H.M., Askanase A.D., et al. Combined oral contraceptives in women with systemic lupus erythematosus. N. Engl. J. Med. 2005;353:2550–2558. doi: 10.1056/NEJMoa051135.
    1. Furie R.A., Petri M.A., Wallace D.J., Ginzler E.M., Merrill J.T., Stohl W., Chatham W.W., Strand V., Weinstein A., Chevrier M.R., et al. Novel evidence-based systemic lupus erythematosus responder index. Arthritis Rheum. 2009;61:1143–1151. doi: 10.1002/art.24698.
    1. Hay E.M., Bacon P.A., Gordon C., Isenberg D.A., Maddison P., Snaith M.L., Symmons D.P., Viner N., Zoma A. The BILAG index: A reliable and valid instrument for measuring clinical disease activity in systemic lupus erythematosus. QJM Int. J. Med. 1993;86:447–458.
    1. Gladman D., Ginzler E., Goldsmith C., Fortin P., Liang M., Urowitz M., Bacon P., Bombardieri S., Hanly J., Hay E., et al. The development and initial validation of the Systemic Lupus International Collaborating Clinics/American College of Rheumatology damage index for systemic lupus erythematosus. Arthritis Rheum. 1996;39:363–369. doi: 10.1002/art.1780390303.
    1. Jacobi A.M., Odendahl M., Reiter K., Bruns A., Burmester G.R., Radbruch A., Valet G., Lipsky P.E., Dorner T. Correlation between circulating CD27high plasma cells and disease activity in patients with systemic lupus erythematosus. Arthritis Rheum. 2003;48:1332–1342. doi: 10.1002/art.10949.
    1. Ellyard J.I., Avery D.T., Phan T.G., Hare N.J., Hodgkin P.D., Tangye S.G. Antigen-selected, immunoglobulin-secreting cells persist in human spleen and bone marrow. Blood. 2004;103:3805–3812. doi: 10.1182/blood-2003-09-3109.
    1. Klasener K., Jellusova J., Andrieux G., Salzer U., Bohler C., Steiner S.N., Albinus J.B., Cavallari M., Suss B., Voll R.E., et al. CD20 as a gatekeeper of the resting state of human B cells. Proc. Natl. Acad. Sci. USA. 2021;118:e2021342118. doi: 10.1073/pnas.2021342118.
    1. Sanz I., Wei C., Jenks S.A., Cashman K.S., Tipton C., Woodruff M.C., Hom J., Lee F.E. Challenges and Opportunities for Consistent Classification of Human B Cell and Plasma Cell Populations. Front. Immunol. 2019;10:2458. doi: 10.3389/fimmu.2019.02458.

Source: PubMed

3
Prenumerera