Repopulation of T, B, and NK cells following alemtuzumab treatment in relapsing-remitting multiple sclerosis

Wendy Gilmore, Brett T Lund, Peili Li, Alex M Levy, Eve E Kelland, Omid Akbari, Susan Groshen, Steven Yong Cen, Daniel Pelletier, Leslie P Weiner, Adil Javed, Jeffrey E Dunn, Anthony L Traboulsee, Wendy Gilmore, Brett T Lund, Peili Li, Alex M Levy, Eve E Kelland, Omid Akbari, Susan Groshen, Steven Yong Cen, Daniel Pelletier, Leslie P Weiner, Adil Javed, Jeffrey E Dunn, Anthony L Traboulsee

Abstract

Objective: To characterize long-term repopulation of peripheral immune cells following alemtuzumab-induced lymphopenia in relapsing-remitting MS (RRMS), with a focus on regulatory cell types, and to explore associations with clinical outcome measures.

Methods: The project was designed as a multicenter add-on longitudinal mechanistic study for RRMS patients enrolled in CARE-MS II, CARE-MS II extension at the University of Southern California and Stanford University, and an investigator-initiated study conducted at the Universities of British Columbia and Chicago. Methods involved collection of blood at baseline, prior to alemtuzumab administration, and at months 5, 11, 17, 23, 36, and 48 post-treatment. T cell, B cell, and natural killer (NK) cell subsets, chemokine receptor expression in T cells, in vitro cytokine secretion patterns, and regulatory T cell (Treg) function were assessed. Clinical outcomes, including expanded disability status score (EDSS), relapses, conventional magnetic resonance imaging (MRI) measures, and incidents of secondary autoimmunity were tracked.

Results: Variable shifts in lymphocyte populations occurred over time in favor of CD4+ T cells, B cells, and NK cells with surface phenotypes characteristic of regulatory subsets, accompanied by reduced ratios of effector to regulatory cell types. Evidence of increased Treg competence was observed after each treatment course. CD4+ and CD8+ T cells that express CXCR3 and CCR5 and CD8+ T cells that express CDR3 and CCR4 were also enriched after treatment, indicating heightened trafficking potential in activated T cells. Patterns of repopulation were not associated with measures of clinical efficacy or secondary autoimmunity, but exploratory analyses using a random generalized estimating equation (GEE) Poisson model provide preliminary evidence of associations between pro-inflammatory cell types and increased risk for gadolinium (Gd+) enhancing lesions, while regulatory subsets were associated with reduced risk. In addition, the risk for T2 lesions correlated with increases in CD3+CD8+CXCR3+ cells.

Conclusions: Lymphocyte repopulation after alemtuzumab treatment favors regulatory subsets in the T cell, B cell, and NK cell compartments. Clinical efficacy may reflect the sum of interactions among them, leading to control of potentially pathogenic effector cell types. Several immune measures were identified as possible biomarkers of lesion activity. Future studies are necessary to more precisely define regulatory and effector subsets and their contributions to clinical efficacy and risk for secondary autoimmunity in alemtuzumab-treated patients, and to reveal new insights into mechanisms of immunopathogenesis in MS.

Trial registration: Parent trials for this study are registered with ClinicalTrials.gov: CARE-MS II: NCT00548405, CARE-MS II extension: NCT00930553 and ISS: NCT01307332.

Keywords: Alemtuzumab; B cell subsets; Drug mechanisms; Immune regulation; Lymphocyte repopulation; Lymphopenia; Multiple sclerosis; Natural killer cell subsets; T cell subsets; Tolerance.

Conflict of interest statement

WG has received funding from Sanofi/Genzyme for investigator-initiated research and honoraria for service on advisory and scientific boards for Sanofi/Genzyme. BTL has received funding from Sanofi/Genzyme, Novartis Pharmaceuticals Corporation, and Teva Pharmaceuticals for investigator-initiated research and has received honoraria from Teva Pharmaceuticals. PL has no disclosures to report. AML has no disclosures to report. EEK has received funding for investigator-initiated research from Novartis Pharmaceuticals Corporation and Teva Pharmaceuticals. OA has no disclosures to report. SG has no disclosures to report. SYP has no disclosures to report. DP has received consultation honoraria from Biogen, EMD Serono, Genentech, Genzyme Corporation, Novartis Pharmaceuticals, and Teva Pharmaceuticals and research funding from Genzyme Corporation. LPW has no disclosures to report. AJ has received consultation honoraria from EMD-Serono, Sanofi-Genzyme, Novartis, Biogen, and Genentech. JED has received honoraria for participation on Advisory Boards from Genentech, Biogen, and Alexion and compensation for education programs from Novartis and Sanofi. ALT has received grant funding from the MS Society of Canada, Canadian Institute for Health Research, Roche, and Genzyme. He has received honoraria or travel grants from Teva Canada Innovation, Roche, Merck/EMD Serono, Genzyme, and Chugai Pharmaceuticals.

Figures

Fig. 1
Fig. 1
Changes in CD4+ regulatory T cells (Treg), “effector” T cells (Teff), and Treg function in PBMC. a FACS gating strategy used to identify CD4 + CD25hiCD127lo/neg foxP3+ Tregs and CD4 + CD25 + CD127 + foxP3- Teff in PBMC (first four FACS plots, left to right) and in PBMC depleted of CD25+ Tregs (fifth FACS plot, right). b-e Illustrate changes over time at each study timepoint for each individual patient (open symbols), compared with baseline (M0) values. b Changes in Tregs. c Changes in Teff. d Changes in Teff:Treg ratio. e Changes in Treg function, indicated as percent rebound in CD25-depleted PBMC compared to undepleted PBMC. Data in all panels represent median/IQR values for all patients at each timepoint. p values indicate significant changes from baseline (M0) as follows: ****p ≤ 0.0001, ***p ≤ 0.001, **p ≤ 0.01, *p ≤ 0.05; mixed effects ANOVA with Tukey’s corrections for multiple comparisons
Fig. 2
Fig. 2
Changes in CD4+ and CD8+ T cells expressing chemokine receptors in whole blood. a Total CD3+CD4+ T lymphocytes. b Total CD3+ CD8+ T cells. c and d Show CD3+CD4+ T cells expressing CXCR3 and CCR5, respectively, which are enriched in Th1 cells. e and f Show CD3+CD4+ T cells expressing CCR3 and CCR4, respectively, which are enriched in Th2 cells. gj Show CD3+CD8+ T cells that express CXCR3, CCR5, CCR3, and CCR4, respectively. Data represent median/IQR values for all patients at each timepoint; open circles indicate data points for each individual patient. p values indicate significant changes from baseline (M0) values as follows: ****p ≤ 0.0001, ***p ≤ 0.001, **p ≤ 0.01, *p ≤ 0.05; mixed effects ANOVA with Tukey’s corrections for multiple comparisons
Fig. 3
Fig. 3
Changes in B cell subsets in thawed PBMC. a FACS gating strategy used to identify CD19+20+CD27+ total memory B cells, CD19+20+ total naïve B cells, and CD19+ CD20+CD27-CD24hiCD38hi B cells. b Total CD19+CD20+CD27+ memory B cells. c CD19+CD20+CD27- total naïve B cells. d CD19+CD20+CD27-CD24hiCD38hi B cells. e Ratio of total memory B cells to naïve “regulatory” CD24hiCD38hi B cells. f Ratio of total naïve B cells to CD19+CD20+CD27-CD24hiCD38hi B cells. Data represent median/IQR values for all patients at each timepoint; open circles indicate data points for each individual patient. p values indicate significant changes from baseline (M0) values as follows: ****p ≤ 0.0001, ***p ≤ 0.001, **p ≤ 0.01, *p ≤ 0.05; Kruskal-Wallis test
Fig. 4
Fig. 4
Changes in NK cell subsets in whole blood. a FACS gating strategy used to identify CD3-CD56+ total NK cells, CD3-CD56bright NK cells, and CD3+CD56+ NKT cells. b Total CD3-CD56+ NK cells. c CD3-CD56bright NK cells. d CD3+ CD56+ NKT cells. e Ratio of NKT cells:CD3-CD56bright T cells. Data represent median/IQR values for all patients at each timepoint; open circles indicate data points for each individual patient. p values indicate significant changes from baseline (M0) values as follows: ****p ≤ 0.0001, ***p ≤ 0.001, **p ≤ 0.01, *p ≤ 0.05; mixed effects ANOVA with Tukey’s corrections for multiple comparisons
Fig. 5
Fig. 5
Clinical and imaging outcomes. a Expanded disability status scores (EDSS), measured every 6 months. b Number of new T2 lesions. c T2 volume. d Number of Gd+ lesions at baseline; subsequent timepoints represent new Gd+ lesions. e Brain parenchymal fraction (BPF). Data are expressed as median/IQR values for all patients at each timepoint. p values indicate significant changes from baseline (M0), where applicable, as follows: **p ≤ 0.01, *p ≤ 0.05; Kruskal-Wallis test for EDSS, number of new T2 and Gd+ lesions, and mixed effects ANOVA with Tukey’s corrections for T2 volume and BPF. f Number of patients with new T2 lesions in the first year (up to M12), second year (M13–M24), third year (M25–M36), and fourth year (M37–M48) accompanied by total number for all timepoints on study. g Total number of new lesions in each year. h Number of patients with Gd+ lesions at M0 [11] and new Gd+ lesions in each year, accompanied by the total number of new Gd+ lesions from M12 to M48. i Total number Gd+ lesions at M0 and new Gd+ lesions in each year. j Number of patients who experienced relapses in each year on study, accompanied by the total for all years. k Number of patients who experienced relapses and/or new Gd+ lesions (active disease) or had no relapses or Gd+ lesions (stable disease) on study
Fig. 6
Fig. 6
Lack of significant differences in selected lymphocyte subsets stratified for patients with active and stable disease activity during the 48-month study period. Each row represents data from one lymphocyte subset, with the left panels in each row illustrating between-subjects analyses, followed by within-subjects analyses for patients with active (middle panels) and stable (right panels) disease activity. ac Changes in CD4+CD25+CD127+foxP3- Teff. df Changes in CD4+CD25hiCD127lo/neg foxP3+ Tregs. gi Changes in CD19+CD20+CD27- total naïve B cells. jl Changes in CD19+CD20+CD27-CD24hi CD38hi B cells. mo Changes in CD3-CD56bright NK cells. pr Changes in CD3+CD56+ NKT cells. Data represent median/IQR; closed symbols indicate patients with active disease, and open symbols indicate patients with stable disease. Data were subjected to between- and within-subject analyses using a linear mixed effects model for repeated measures
Fig. 7
Fig. 7
Lack of significant differences in selected lymphocyte subsets stratified for patients with and without secondary autoimmunity during the 48-month study period. Each row represents data from one lymphocyte subset, with the left panels in each row illustrating between-subjects analyses, followed by within-subjects analyses for patients with secondary autoimmunity (middle panels) and without secondary autoimmunity (right panels) disease activity. ac Changes in CD4+CD25+CD127+foxP3- Teff. df Changes in CD4+CD25hiCD127lo/neg foxP3+ Tregs. gi Changes in CD19+CD20+CD27- total naïve B cells. jl Changes in CD19+CD20+CD27-CD24hi CD38hi B cells. mo Changes in CD3-CD56bright NK cells. pr Changes in CD3+CD56+ NKT cells. Data represent median/IQR; closed symbols indicate patients with secondary autoimmunity, and open symbols indicate patients without secondary autoimmunity. Differences over time between and within groups were not statistically significant (linear mixed effects model for repeated measures)

References

    1. Martin R, Sospedra M, Rosito M, Engelhardt B. Current multiple sclerosis treatments have improved our understanding of MS autoimmune pathogenesis. Eur J Immunol. 2016;46(9):2078–2090.
    1. Baecher-Allan C, Kaskow BJ, Weiner HL. Multiple sclerosis: mechanisms and immunotherapy. Neuron. 2018;97(4):742–768.
    1. Luckel C, Picard F, Raifer H, Campos Carrascosa L, Guralnik A, Zhang Y, et al. IL-17(+) CD8(+) T cell suppression by dimethyl fumarate associates with clinical response in multiple sclerosis. Nat Commun. 2019;10(1):5722.
    1. Sabatino JJ, Jr, Wilson MR, Calabresi PA, Hauser SL, Schneck JP, Zamvil SS. Anti-CD20 therapy depletes activated myelin-specific CD8(+) T cells in multiple sclerosis. Proc Natl Acad Sci U S A. 2019;116(51):25800–25807.
    1. Bielekova B, Catalfamo M, Reichert-Scrivner S, Packer A, Cerna M, Waldmann TA, et al. Regulatory CD56(bright) natural killer cells mediate immunomodulatory effects of IL-2Ralpha-targeted therapy (daclizumab) in multiple sclerosis. Proc Natl Acad Sci U S A. 2006;103(15):5941–5946.
    1. Prod'homme T, Zamvil SS. The evolving mechanisms of action of glatiramer acetate. Cold Spring Harb Perspect Med. 2019;9:2.
    1. Hauser SL. The Charcot Lecture | beating MS: a story of B cells, with twists and turns. Mult Scler. 2015;21(1):8–21.
    1. Moreno Torres I, Garcia-Merino A. Anti-CD20 monoclonal antibodies in multiple sclerosis. Expert Rev Neurother. 2017;17(4):359–371.
    1. Lehmann-Horn K, Kinzel S, Weber MS. Deciphering the role of B cells in multiple sclerosis-towards specific targeting of pathogenic function. Int J Mol Sci. 2017;18:10.
    1. Grutzke B, Hucke S, Gross CC, Herold MV, Posevitz-Fejfar A, Wildemann BT, et al. Fingolimod treatment promotes regulatory phenotype and function of B cells. Ann Clin Transl Neurol. 2015;2(2):119–130.
    1. Baker D, Herrod SS, Alvarez-Gonzalez C, Zalewski L, Albor C, Schmierer K. Both cladribine and alemtuzumab may effect MS via B-cell depletion. Neurol Neuroimmunol Neuroinflamm. 2017;4(4):e360.
    1. Kuerten S, Jackson LJ, Kaye J, Vollmer TL. Impact of glatiramer acetate on B cell-mediated pathogenesis of multiple sclerosis. CNS Drugs. 2018;32(11):1039–1051.
    1. Longbrake EE, Cross AH. Effect of multiple sclerosis disease-modifying therapies on B cells and humoral immunity. JAMA Neurol. 2016;73(2):219–225.
    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. Cohen JA, Coles AJ, Arnold DL, Confavreux C, Fox EJ, Hartung HP, et al. Alemtuzumab versus interferon beta 1a as first-line treatment for patients with relapsing-remitting multiple sclerosis: a randomised controlled phase 3 trial. Lancet. 2012;380(9856):1819–1828.
    1. Coles AJ, Twyman CL, Arnold DL, Cohen JA, Confavreux C, Fox EJ, et al. Alemtuzumab for patients with relapsing multiple sclerosis after disease-modifying therapy: a randomised controlled phase 3 trial. Lancet. 2012;380(9856):1829–1839.
    1. Investigators CT, Coles AJ, Compston DA, Selmaj KW, Lake SL, Moran S, et al. Alemtuzumab vs. interferon beta-1a in early multiple sclerosis. N Engl J Med. 2008;359(17):1786–1801.
    1. Katsavos S, Coles A. Alemtuzumab as treatment for multiple sclerosis. Cold Spring Harb Perspect Med. 2018;8:10.
    1. Coles AJ, Cohen JA, Fox EJ, Giovannoni G, Hartung HP, Havrdova E, et al. Alemtuzumab CARE-MS II 5-year follow-up: efficacy and safety findings. Neurology. 2017;89(11):1117–1126.
    1. Ambrose LR, Morel AS, Warrens AN. Neutrophils express CD52 and exhibit complement-mediated lysis in the presence of alemtuzumab. Blood. 2009;114(14):3052–3055.
    1. Gross CC, Ahmetspahic D, Ruck T, Schulte-Mecklenbeck A, Schwarte K, Jorgens S, et al. Alemtuzumab treatment alters circulating innate immune cells in multiple sclerosis. Neurol Neuroimmunol Neuroinflamm. 2016;3(6):e289.
    1. Irimata K, Wakim P, Li X. Estimation of correlation coefficient in data with repeated measures. SAS Global Proceedings. 2018;.
    1. Cox AL, Thompson SA, Jones JL, Robertson VH, Hale G, Waldmann H, et al. Lymphocyte homeostasis following therapeutic lymphocyte depletion in multiple sclerosis. Eur J Immunol. 2005;35(11):3332–3342.
    1. Freedman MS, Kaplan JM, Markovic-Plese S. Insights into the mechanisms of the therapeutic efficacy of alemtuzumab in multiple sclerosis. J Clin Cell Immunol. 2013;4:4.
    1. Ziemssen T, Thomas K. Alemtuzumab in the long-term treatment of relapsing-remitting multiple sclerosis: an update on the clinical trial evidence and data from the real world. Ther Adv Neurol Disord. 2017;10(10):343–359.
    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(8):961–969.
    1. Hill-Cawthorne GA, Button T, Tuohy O, Jones JL, May K, Somerfield J, et al. Long term lymphocyte reconstitution after alemtuzumab treatment of multiple sclerosis. J Neurol Neurosurg Psychiatry. 2012;83(3):298–304.
    1. Jones JL, Coles AJ. Mode of action and clinical studies with alemtuzumab. Exp Neurol. 2014;262 Pt A:37-43.
    1. De Mercanti S, Rolla S, Cucci A, Bardina V, Cocco E, Vladic A, et al. Alemtuzumab long-term immunologic effect: Treg suppressor function increases up to 24 months. Neurol Neuroimmunol Neuroinflamm. 2016;3(1):e194.
    1. Zhang X, Tao Y, Chopra M, Ahn M, Marcus KL, Choudhary N, et al. Differential reconstitution of T cell subsets following immunodepleting treatment with alemtuzumab (anti-CD52 monoclonal antibody) in patients with relapsing-remitting multiple sclerosis. J Immunol. 2013;191(12):5867–5874.
    1. Heidt S, Hester J, Shankar S, Friend PJ, Wood KJ. B cell repopulation after alemtuzumab induction-transient increase in transitional B cells and long-term dominance of naive B cells. Am J Transplant. 2012;12(7):1784–1792.
    1. Vavasour IM, Tam R, Li DK, Laule C, Taylor C, Kolind SH, et al. A 24-month advanced magnetic resonance imaging study of multiple sclerosis patients treated with alemtuzumab. Mult Scler. 2018:1352458518770085.
    1. Vavasour IM, Tam R, Li DK, Laule C, Taylor C, Kolind SH, et al. A 24-month advanced magnetic resonance imaging study of multiple sclerosis patients treated with alemtuzumab. Mult Scler. 2019;25(6):811–818.
    1. Lund BT, Kelland EE, Hayardeny L, Barilan O, Gilmore W, Weiner LP. Assessment of changes in immune measures of multiple sclerosis patients treated with laquinimod. J Neuroimmunol. 2013;263(1-2):108–115.
    1. Lovett-Racke AE, Gormley M, Liu Y, Yang Y, Graham C, Wray S, et al. B cell depletion with ublituximab reshapes the T cell profile in multiple sclerosis patients. J Neuroimmunol. 2019;332:187–197.
    1. Gilmore W, Arias M, Stroud N, Stek A, McCarthy KA, Correale J. Preliminary studies of cytokine secretion patterns associated with pregnancy in MS patients. J Neurol Sci. 2004;224(1-2):69–76.
    1. Jones JL, Thompson SA, Loh P, Davies JL, Tuohy OC, Curry AJ, et al. Human autoimmunity after lymphocyte depletion is caused by homeostatic T-cell proliferation. Proc Natl Acad Sci U S A. 2013;110(50):20200–20205.
    1. Santegoets SJ, Dijkgraaf EM, Battaglia A, Beckhove P, Britten CM, Gallimore A, et al. Monitoring regulatory T cells in clinical samples: consensus on an essential marker set and gating strategy for regulatory T cell analysis by flow cytometry. Cancer Immunol Immunother. 2015;64(10):1271–1286.
    1. Kim Y, Kim G, Shin HJ, Hyun JW, Kim SH, Lee E, et al. Restoration of regulatory B cell deficiency following alemtuzumab therapy in patients with relapsing multiple sclerosis. J Neuroinflammation. 2018;15(1):300.
    1. McMurchy AN, Levings MK. Suppression assays with human T regulatory cells: a technical guide. Eur J Immunol. 2012;42(1):27–34.
    1. Baecher-Allan CM, Hafler DA. The purification and functional analysis of human CD4 + CD25high regulatory T cells. Curr Protoc Immunol. 2006;Chapter 7:Unit 7 4B.
    1. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155(3):1151–1164.
    1. Engler JB, Undeutsch R, Kloke L, Rosenberger S, Backhaus M, Schneider U, et al. Unmasking of autoreactive CD4 T cells by depletion of CD25 regulatory T cells in systemic lupus erythematosus. Ann Rheum Dis. 2011;70(12):2176–2183.
    1. Lund BT, Ashikian N, Ta HQ, Chakryan Y, Manoukian K, Groshen S, et al. Increased CXCL8 (IL-8) expression in multiple sclerosis. J Neuroimmunol. 2004;155(1-2):161–171.
    1. Rivino L, Messi M, Jarrossay D, Lanzavecchia A, Sallusto F, Geginat J. Chemokine receptor expression identifies Pre-T helper (Th)1, Pre-Th2, and nonpolarized cells among human CD4+ central memory T cells. J Exp Med. 2004;200(6):725–735.
    1. Kunicki MA, Amaya Hernandez LC, Davis KL, Bacchetta R, Roncarolo MG. Identity and diversity of human peripheral Th and T regulatory cells defined by single-cell mass cytometry. J Immunol. 2018;200(1):336–346.
    1. Annunziato F, Cosmi L, Galli G, Beltrame C, Romagnani P, Manetti R, et al. Assessment of chemokine receptor expression by human Th1 and Th2 cells in vitro and in vivo. J Leukoc Biol. 1999;65(5):691–699.
    1. Fuhrman CA, Yeh WI, Seay HR, Saikumar Lakshmi P, Chopra G, Zhang L, et al. Divergent phenotypes of human regulatory T cells expressing the receptors TIGIT and CD226. J Immunol. 2015;195(1):145–155.
    1. Nolz JC. Molecular mechanisms of CD8(+) T cell trafficking and localization. Cell Mol Life Sci. 2015;72(13):2461–2473.
    1. Groom JR, Luster AD. CXCR3 in T cell function. Exp Cell Res. 2011;317(5):620–631.
    1. Sospedra M, Martin R. Immunology of multiple sclerosis. Semin Neurol. 2016;36(2):115–127.
    1. von Budingen HC, Palanichamy A, Lehmann-Horn K, Michel BA, Zamvil SS. Update on the autoimmune pathology of multiple sclerosis: B-cells as disease-drivers and therapeutic targets. Eur Neurol. 2015;73(3-4):238–246.
    1. Greenfield AL, Hauser SL. B-cell therapy for multiple sclerosis: entering an era. Ann Neurol. 2018;83(1):13–26.
    1. Cross AH, Waubant E. MS and the B cell controversy. Biochim Biophys Acta. 2011;1812(2):231–238.
    1. Andre P, Anfossi N. Clinical analysis of human natural killer cells. Methods Mol Biol. 2008;415:291–300.
    1. Thompson SA, Jones JL, Cox AL, Compston DA, Coles AJ. B-cell reconstitution and BAFF after alemtuzumab (Campath-1H) treatment of multiple sclerosis. J Clin Immunol. 2010;30(1):99–105.
    1. Coles AJ, Cox A, Le Page E, Jones J, Trip SA, Deans J, et al. The window of therapeutic opportunity in multiple sclerosis: evidence from monoclonal antibody therapy. J Neurol. 2006;253(1):98–108.
    1. Ruck T, Bittner S, Wiendl H, Meuth SG. Alemtuzumab in multiple sclerosis: mechanism of action and beyond. Int J Mol Sci. 2015;16(7):16414–16439.
    1. Coles AJ, Wing M, Smith S, Coraddu F, Greer S, Taylor C, et al. Pulsed monoclonal antibody treatment and autoimmune thyroid disease in multiple sclerosis. Lancet. 1999;354(9191):1691–1695.
    1. Antonioli L, Pacher P, Vizi ES, Hasko G. CD39 and CD73 in immunity and inflammation. Trends Mol Med. 2013;19(6):355–367.
    1. Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol Rev. 2017;276(1):121–144.
    1. Dwyer KM, Deaglio S, Gao W, Friedman D, Strom TB, Robson SC. CD39 and control of cellular immune responses. Purinergic Signal. 2007;3(1-2):171–180.
    1. Pant AB, Wang Y, Mielcarz DW, Kasper EJ, Telesford KM, Mishra M, et al. Alteration of CD39 + Foxp3+ CD4 T cell and cytokine levels in EAE/MS following anti-CD52 treatment. J Neuroimmunol. 2017;303:22–30.
    1. Borsellino G, Kleinewietfeld M, Di Mitri D, Sternjak A, Diamantini A, Giometto R, et al. Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood. 2007;110(4):1225–1232.
    1. Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204(6):1257–1265.
    1. Dwyer KM, Hanidziar D, Putheti P, Hill PA, Pommey S, McRae JL, et al. Expression of CD39 by human peripheral blood CD4+ CD25+ T cells denotes a regulatory memory phenotype. Am J Transplant. 2010;10(11):2410–2420.
    1. Gu J, Ni X, Pan X, Lu H, Lu Y, Zhao J, et al. Human CD39(hi) regulatory T cells present stronger stability and function under inflammatory conditions. Cell Mol Immunol. 2017;14(6):521–528.
    1. Dalla Libera D, Di Mitri D, Bergami A, Centonze D, Gasperini C, Grasso MG, et al. T regulatory cells are markers of disease activity in multiple sclerosis patients. PLoS One. 2011;6(6):e21386.
    1. Fletcher JM, Lonergan R, Costelloe L, Kinsella K, Moran B, O'Farrelly C, et al. CD39 + Foxp3+ regulatory T cells suppress pathogenic Th17 cells and are impaired in multiple sclerosis. J Immunol. 2009;183(11):7602–7610.
    1. Kitz A, Singer E, Hafler D. Regulatory T cells: from discovery to autoimmunity. Cold Spring Harb Perspect Med. 2018;8:12.
    1. Viglietta V, Baecher-Allan C, Weiner HL, Hafler DA. Loss of functional suppression by CD4 + CD25+ regulatory T cells in patients with multiple sclerosis. J Exp Med. 2004;199(7):971–979.
    1. Weingartner E, Golding A. Direct control of B cells by Tregs: an opportunity for long-term modulation of the humoral response. Cell Immunol. 2017;318:8–16.
    1. Flores-Borja F, Bosma A, Ng D, Reddy V, Ehrenstein MR, Isenberg DA, et al. CD19 + CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation. Sci Transl Med. 2013;5(173):173ra23.
    1. Mauri C, Menon M. Human regulatory B cells in health and disease: therapeutic potential. J Clin Invest. 2017;127(3):772–779.
    1. Vacca P, Cantoni C, Vitale M, Prato C, Canegallo F, Fenoglio D, et al. Crosstalk between decidual NK and CD14+ myelomonocytic cells results in induction of Tregs and immunosuppression. Proc Natl Acad Sci U S A. 2010;107(26):11918–11923.
    1. La Cava A, Van Kaer L, Fu DS. CD4 + CD25+ Tregs and NKT cells: regulators regulating regulators. Trends Immunol. 2006;27(7):322–327.
    1. Van Kaer L, Wu L, Parekh VV. Natural killer T cells in multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis. Immunology. 2015;146(1):1–10.
    1. Gianchecchi E, Delfino DV, Fierabracci A. NK cells in autoimmune diseases: linking innate and adaptive immune responses. Autoimmun Rev. 2018;17(2):142–154.
    1. Schuster IS, Coudert JD, Andoniou CE, Degli-Esposti MA. “Natural Regulators”: NK cells as modulators of T cell immunity. Front Immunol. 2016;7:235.
    1. Nakagawa H, Wang L, Cantor H, Kim HJ. New insights into the biology of CD8 regulatory T cells. Adv Immunol. 2018;140:1–20.
    1. Wiendl H, Carraro M, Comi G, Izquierdo G, Kim HJ, Sharrack B, et al. Lymphocyte pharmacodynamics are not associated with autoimmunity or efficacy after alemtuzumab. Neurology(R) neuroimmunology & neuroinflammation. 2020;7:1.
    1. Jones JL, Anderson JM, Phuah CL, Fox EJ, Selmaj K, Margolin D, et al. Improvement in disability after alemtuzumab treatment of multiple sclerosis is associated with neuroprotective autoimmunity. Brain. 2010;133(Pt 8):2232–2247.
    1. Jones JL, Phuah CL, Cox AL, Thompson SA, Ban M, Shawcross J, et al. IL-21 drives secondary autoimmunity in patients with multiple sclerosis, following therapeutic lymphocyte depletion with alemtuzumab (Campath-1H) J Clin Invest. 2009;119(7):2052–2061.
    1. Bitsch A, Schuchardt J, Bunkowski S, Kuhlmann T, Bruck W. Acute axonal injury in multiple sclerosis. Correlation with demyelination and inflammation. Brain. 2000;123(Pt 6):1174–1183.
    1. Booss J, Esiri MM, Tourtellotte WW, Mason DY. Immunohistological analysis of T lymphocyte subsets in the central nervous system in chronic progressive multiple sclerosis. J Neurol Sci. 1983;62(1-3):219–232.
    1. Friese MA, Fugger L. Pathogenic CD8(+) T cells in multiple sclerosis. Ann Neurol. 2009;66(2):132–141.
    1. Hauser SL, Bhan AK, Gilles F, Kemp M, Kerr C, Weiner HL. Immunohistochemical analysis of the cellular infiltrate in multiple sclerosis lesions. Ann Neurol. 1986;19(6):578–587.
    1. Milo R. The efficacy and safety of daclizumab and its potential role in the treatment of multiple sclerosis. Ther Adv Neurol Disord. 2014;7(1):7–21.
    1. Michel T, Poli A, Cuapio A, Briquemont B, Iserentant G, Ollert M, et al. Human CD56bright NK cells: an update. J Immunol. 2016;196(7):2923–2931.
    1. Kaskow BJ, Baecher-Allan C. Effector T cells in multiple sclerosis. Cold Spring Harb Perspect Med. 2018;8:4.
    1. Massey JC, Sutton IJ, Ma DDF, Moore JJ. Regenerating immunotolerance in multiple sclerosis with autologous hematopoietic stem cell transplant. Front Immunol. 2018;9:410.
    1. Bloom DD, Chang Z, Fechner JH, Dar W, Polster SP, Pascual J, et al. CD4+ CD25+ FOXP3+ regulatory T cells increase de novo in kidney transplant patients after immunodepletion with Campath-1H. Am J Transplant. 2008;8(4):793–802.
    1. Simon AK, Hollander GA, McMichael A. Evolution of the immune system in humans from infancy to old age. Proc Biol Sci. 2015;282(1821):20143085.

Source: PubMed

3
Prenumerera