Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2018

Open Access 01-12-2018 | Research

Anti-CD52 antibody treatment depletes B cell aggregates in the central nervous system in a mouse model of multiple sclerosis

Authors: Micha Simon, Rojda Ipek, György A. Homola, Damiano M. Rovituso, Andrea Schampel, Christoph Kleinschnitz, Stefanie Kuerten

Published in: Journal of Neuroinflammation | Issue 1/2018

Login to get access

Abstract

Background

Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) for which several new treatment options were recently introduced. Among them is the monoclonal anti-CD52 antibody alemtuzumab that depletes mainly B cells and T cells in the immune periphery. Considering the ongoing controversy about the involvement of B cells and in particular the formation of B cell aggregates in the brains of progressive MS patients, an in-depth understanding of the effects of anti-CD52 antibody treatment on the B cell compartment in the CNS itself is desirable.

Methods

We used myelin basic protein (MBP)-proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE) in C57BL/6 (B6) mice as B cell-dependent model of MS. Mice were treated intraperitoneally either at the peak of EAE or at 60 days after onset with 200 μg murine anti-CD52 vs. IgG2a isotype control antibody for five consecutive days. Disease was subsequently monitored for 10 days. The antigen-specific B cell/antibody response was measured by ELISPOT and ELISA. Effects on CNS infiltration and B cell aggregation were determined by immunohistochemistry. Neurodegeneration was evaluated by Luxol Fast Blue, SMI-32, and Olig2/APC staining as well as by electron microscopy and phosphorylated heavy neurofilament serum ELISA.

Results

Treatment with anti-CD52 antibody attenuated EAE only when administered at the peak of disease. While there was no effect on the production of MP4-specific IgG, the treatment almost completely depleted CNS infiltrates and B cell aggregates even when given as late as 60 days after onset. On the ultrastructural level, we observed significantly less axonal damage in the spinal cord and cerebellum in chronic EAE after anti-CD52 treatment.

Conclusion

Anti-CD52 treatment abrogated B cell infiltration and disrupted existing B cell aggregates in the CNS.
Appendix
Available only for authorised users
Literature
2.
go back to reference Popescu BF, Pirko I, Lucchinetti CF. Pathology of multiple sclerosis: where do we stand? Continuum (Minneap Minn). 2013;19:901–21. Popescu BF, Pirko I, Lucchinetti CF. Pathology of multiple sclerosis: where do we stand? Continuum (Minneap Minn). 2013;19:901–21.
3.
go back to reference Disanto G, Morahan JM, Barnett MH, Giovannoni G, Ramagopalan SV. The evidence for a role of B cells in multiple sclerosis. Neurology. 2012;78:823–32.CrossRefPubMedPubMedCentral Disanto G, Morahan JM, Barnett MH, Giovannoni G, Ramagopalan SV. The evidence for a role of B cells in multiple sclerosis. Neurology. 2012;78:823–32.CrossRefPubMedPubMedCentral
4.
go back to reference Lucchinetti C, Bruck W, Parisi J, Scheithauer B, Rodriguez M, Lassmann H. Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann Neurol. 2000;47:707–17.CrossRefPubMed Lucchinetti C, Bruck W, Parisi J, Scheithauer B, Rodriguez M, Lassmann H. Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann Neurol. 2000;47:707–17.CrossRefPubMed
5.
go back to reference Meinl E, Krumbholz M, Hohlfeld R. B lineage cells in the inflammatory central nervous system environment: migration, maintenance, local antibody production, and therapeutic modulation. Ann Neurol. 2006;59:880–92.CrossRefPubMed Meinl E, Krumbholz M, Hohlfeld R. B lineage cells in the inflammatory central nervous system environment: migration, maintenance, local antibody production, and therapeutic modulation. Ann Neurol. 2006;59:880–92.CrossRefPubMed
6.
go back to reference Hauser SL, Bar-Or A, Comi G, Giovannoni G, Hartung HP, Hemmer B, Lublin F, Montalban X, Rammohan KW, Selmaj K, Traboulsee A, Wolinsky JS, Arnold DL, Klingelschmitt G, Masterman D, Fontoura P, Belachew S, Chin P, Mairon N, Garren H, Kappos L, OPERA I, OPERA II Clinical Investigators. Ocrelizumab versus inteferon Beta-1a in relapsing multiple sclerosis. N Engl J Med. 2017;376:221–34.CrossRefPubMed Hauser SL, Bar-Or A, Comi G, Giovannoni G, Hartung HP, Hemmer B, Lublin F, Montalban X, Rammohan KW, Selmaj K, Traboulsee A, Wolinsky JS, Arnold DL, Klingelschmitt G, Masterman D, Fontoura P, Belachew S, Chin P, Mairon N, Garren H, Kappos L, OPERA I, OPERA II Clinical Investigators. Ocrelizumab versus inteferon Beta-1a in relapsing multiple sclerosis. N Engl J Med. 2017;376:221–34.CrossRefPubMed
7.
go back to reference Cortese I, Chaudhry V, So YT, Cantor F, Cornblath DR, Rae-Grant A. Evidence-based guideline update: Plasmapheresis in neurologic disorders: report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology. 2011;3:294–300.CrossRef Cortese I, Chaudhry V, So YT, Cantor F, Cornblath DR, Rae-Grant A. Evidence-based guideline update: Plasmapheresis in neurologic disorders: report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology. 2011;3:294–300.CrossRef
8.
go back to reference Hale G, Xia MQ, Tighe HP, Dyer MJ, Waldmann H. The CAMPATH-1 antigen (CDw52). Tissue Antigens. 1990;35:118–27.CrossRefPubMed Hale G, Xia MQ, Tighe HP, Dyer MJ, Waldmann H. The CAMPATH-1 antigen (CDw52). Tissue Antigens. 1990;35:118–27.CrossRefPubMed
10.
go back to reference Rao SP, Sancho J, Campos-Rivera J, et al. Human peripheral blood mononuclear cells exhibit heterogeneous CD52 expression levels and show differential sensitivity to alemtuzumab mediated cytolysis. PLoS One. 2012;7:e39416.CrossRefPubMedPubMedCentral Rao SP, Sancho J, Campos-Rivera J, et al. Human peripheral blood mononuclear cells exhibit heterogeneous CD52 expression levels and show differential sensitivity to alemtuzumab mediated cytolysis. PLoS One. 2012;7:e39416.CrossRefPubMedPubMedCentral
11.
go back to reference Wiendl H, Bourdette D, Ciccarelli O. Can immune reprogramming with alemtuzumab induce permanent remission in multiple sclerosis? Neurology. 2017;89:1098–100.CrossRefPubMed Wiendl H, Bourdette D, Ciccarelli O. Can immune reprogramming with alemtuzumab induce permanent remission in multiple sclerosis? Neurology. 2017;89:1098–100.CrossRefPubMed
12.
go back to reference Cox AL, Thompson SA, Jones JL, et al. Lymphocyte homeostasis following therapeutic lymphocyte depletion in multiple sclerosis. Eur J Immunol. 2005;35:3332–42.CrossRefPubMed Cox AL, Thompson SA, Jones JL, et al. Lymphocyte homeostasis following therapeutic lymphocyte depletion in multiple sclerosis. Eur J Immunol. 2005;35:3332–42.CrossRefPubMed
13.
go back to reference Zhang X, Tao Y, Chopra M, 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;191:5867–74.CrossRefPubMed Zhang X, Tao Y, Chopra M, 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;191:5867–74.CrossRefPubMed
14.
go back to reference Havari E, Turner MJ, Campos-Rivera J, et al. Impact of alemtuzumab treatment on the survival and function of human regulatory T cells in vitro. Immunology. 2014;141:123–31.CrossRefPubMed Havari E, Turner MJ, Campos-Rivera J, et al. Impact of alemtuzumab treatment on the survival and function of human regulatory T cells in vitro. Immunology. 2014;141:123–31.CrossRefPubMed
15.
go back to reference 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;30:99–105.CrossRefPubMed 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;30:99–105.CrossRefPubMed
16.
go back to reference 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;12:1784–92.CrossRefPubMedPubMedCentral 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;12:1784–92.CrossRefPubMedPubMedCentral
17.
go back to reference Hu Y, Turner MJ, Shields J, et al. Investigation of the mechanism of action of alemtuzumab in a human CD52 transgenic mouse model. Immunology. 2009;128:260–70.CrossRefPubMedPubMedCentral Hu Y, Turner MJ, Shields J, et al. Investigation of the mechanism of action of alemtuzumab in a human CD52 transgenic mouse model. Immunology. 2009;128:260–70.CrossRefPubMedPubMedCentral
18.
go back to reference Turner MJ, Lamorte MJ, Chretien N, et al. Immune status following alemtuzumab treatment in human CD52 transgenic mice. J Neuroimmunol. 2013;261:29–36.CrossRefPubMed Turner MJ, Lamorte MJ, Chretien N, et al. Immune status following alemtuzumab treatment in human CD52 transgenic mice. J Neuroimmunol. 2013;261:29–36.CrossRefPubMed
19.
go back to reference Turner MJ, Pang PT, Chretien N, et al. Reduction of inflammation and preservation of neurological function by anti-CD52 therapy in murine experimental autoimmune encephalomyelitis. J Neuroimmunol. 2015;285:4–12.CrossRefPubMed Turner MJ, Pang PT, Chretien N, et al. Reduction of inflammation and preservation of neurological function by anti-CD52 therapy in murine experimental autoimmune encephalomyelitis. J Neuroimmunol. 2015;285:4–12.CrossRefPubMed
20.
go back to reference Pant AB, Wang Y, Mielcarz DW, Kasper EJ, Telesford KM, Mishra M, Haque A, Channon JY, Kasper LH, Begum-Haque S. Alteration of CD39+Foxp3+ CD4 T cell and cytokine levels in EAE/MS following anti-CD52 treatment. J Neuroimmunol. 2017;303:22–30.CrossRefPubMed Pant AB, Wang Y, Mielcarz DW, Kasper EJ, Telesford KM, Mishra M, Haque A, Channon JY, Kasper LH, Begum-Haque S. Alteration of CD39+Foxp3+ CD4 T cell and cytokine levels in EAE/MS following anti-CD52 treatment. J Neuroimmunol. 2017;303:22–30.CrossRefPubMed
21.
go back to reference Barbour M, Wood R, Hridi SU, Wilson C, McKay G, Bushell TJ, Jiang HR. The therapeutic effect of anti-CD52 treatment in murine experimental autoimmune encephalomyelitis is associated with altered IL-33 and ST2 expression levels. J Neuroimmunol. 2018;318:87–96.CrossRefPubMed Barbour M, Wood R, Hridi SU, Wilson C, McKay G, Bushell TJ, Jiang HR. The therapeutic effect of anti-CD52 treatment in murine experimental autoimmune encephalomyelitis is associated with altered IL-33 and ST2 expression levels. J Neuroimmunol. 2018;318:87–96.CrossRefPubMed
22.
go back to reference Kuerten S, Lichtenegger FS, Faas S, Angelov DN, Tary-Lehmann M, Lehmann PV. MBP-PLP fusion protein-induced EAE in C57BL/6 mice. J Neuroimmunol. 2006;177:99–111.CrossRefPubMed Kuerten S, Lichtenegger FS, Faas S, Angelov DN, Tary-Lehmann M, Lehmann PV. MBP-PLP fusion protein-induced EAE in C57BL/6 mice. J Neuroimmunol. 2006;177:99–111.CrossRefPubMed
23.
go back to reference Kuerten S, Schickel A, Kerkloh C, et al. Tertiary lymphoid organ development coincides with determinant spreading of the myelin-specific T cell response. Acta Neuropathol. 2012;124:861–73.CrossRefPubMed Kuerten S, Schickel A, Kerkloh C, et al. Tertiary lymphoid organ development coincides with determinant spreading of the myelin-specific T cell response. Acta Neuropathol. 2012;124:861–73.CrossRefPubMed
24.
go back to reference Kuerten S, Gruppe TL, Laurentius LM, et al. Differential patterns of spinal cord pathology induced by MP4, MOG peptide 35-55, and PLP peptide 178-191 in C57BL/6 mice. APMIS. 2011;119:336–46.CrossRefPubMed Kuerten S, Gruppe TL, Laurentius LM, et al. Differential patterns of spinal cord pathology induced by MP4, MOG peptide 35-55, and PLP peptide 178-191 in C57BL/6 mice. APMIS. 2011;119:336–46.CrossRefPubMed
25.
go back to reference Kuerten S, Kostova-Bales DA, Frenzel LP, Tigno JT, Tary-Lehmann M, Angelov DN, Lehmann PV. MP4- and MOG:35-55-induced EAE in C57BL/6 mice differentially targets brain, spinal cord and cerebellum. J Neuroimmunol. 2007;189:31–40.CrossRefPubMedPubMedCentral Kuerten S, Kostova-Bales DA, Frenzel LP, Tigno JT, Tary-Lehmann M, Angelov DN, Lehmann PV. MP4- and MOG:35-55-induced EAE in C57BL/6 mice differentially targets brain, spinal cord and cerebellum. J Neuroimmunol. 2007;189:31–40.CrossRefPubMedPubMedCentral
26.
go back to reference Shaw G. Brain neurotrauma: molecular, neuropsychological, and rehabilitation aspects. In: Kobeissy FH, editor. Chapter 21. Frontiers in Neuroengineering. Boca Raton: CRC Press/Taylor & Francis; 2015. Shaw G. Brain neurotrauma: molecular, neuropsychological, and rehabilitation aspects. In: Kobeissy FH, editor. Chapter 21. Frontiers in Neuroengineering. Boca Raton: CRC Press/Taylor & Francis; 2015.
27.
go back to reference Shaw G, Yang C, Ellis R, et al. Hyperphosphorylated neurofilament NF-H is a serum biomarker of axonal injury. Biochem Biophys Res Commun. 2005;336:1268–77.CrossRefPubMed Shaw G, Yang C, Ellis R, et al. Hyperphosphorylated neurofilament NF-H is a serum biomarker of axonal injury. Biochem Biophys Res Commun. 2005;336:1268–77.CrossRefPubMed
28.
go back to reference Lu CH, Macdonald-Wallis C, Gray E, et al. Neurofilament light chain: a prognostic biomarker in amyotrophic lateral sclerosis. Neurology. 2015;84:2247–57.CrossRefPubMedPubMedCentral Lu CH, Macdonald-Wallis C, Gray E, et al. Neurofilament light chain: a prognostic biomarker in amyotrophic lateral sclerosis. Neurology. 2015;84:2247–57.CrossRefPubMedPubMedCentral
29.
go back to reference Li Y, Zhang L, Kallakuri S, Cohen A, Cavanaugh JM. Correlation of mechanical impact responses and biomarker levels: a new model for biomarker evaluation in TBI. J Neurol Sci. 2015;359:280–6.CrossRefPubMed Li Y, Zhang L, Kallakuri S, Cohen A, Cavanaugh JM. Correlation of mechanical impact responses and biomarker levels: a new model for biomarker evaluation in TBI. J Neurol Sci. 2015;359:280–6.CrossRefPubMed
30.
31.
go back to reference Gresle MM, Liu Y, Dagley LF, et al. Serum phosphorylated neurofilament-heavy chain levels in multiple sclerosis patients. J Neurol Neurosurg Psychiatry. 2014;85:1209–13.CrossRefPubMed Gresle MM, Liu Y, Dagley LF, et al. Serum phosphorylated neurofilament-heavy chain levels in multiple sclerosis patients. J Neurol Neurosurg Psychiatry. 2014;85:1209–13.CrossRefPubMed
32.
go back to reference Cohen JA, Coles AJ, Arnold DL, 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;380:1819–28.CrossRefPubMed Cohen JA, Coles AJ, Arnold DL, 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;380:1819–28.CrossRefPubMed
33.
go back to reference Investigators CT, Coles AJ, Compston DA, et al. Alemtuzumab vs. interferon beta-1a in early multiple sclerosis. N Engl J Med. 2008;359:1786–801.CrossRef Investigators CT, Coles AJ, Compston DA, et al. Alemtuzumab vs. interferon beta-1a in early multiple sclerosis. N Engl J Med. 2008;359:1786–801.CrossRef
34.
go back to reference Willis MD, Harding KE, Pickersgill TP, et al. Alemtuzumab for multiple sclerosis: long term follow-up in a multi-centre cohort. Mult Scler. 2016;22:1215–23.CrossRefPubMed Willis MD, Harding KE, Pickersgill TP, et al. Alemtuzumab for multiple sclerosis: long term follow-up in a multi-centre cohort. Mult Scler. 2016;22:1215–23.CrossRefPubMed
35.
go back to reference Havrdova E, Arnold DL, Cohen JA, et al. Alemtuzumab CARE-MS I 5-year follow-up: durable efficacy in the absence of continuous MS therapy. Neurology. 2017;89:1107–16.CrossRefPubMedPubMedCentral Havrdova E, Arnold DL, Cohen JA, et al. Alemtuzumab CARE-MS I 5-year follow-up: durable efficacy in the absence of continuous MS therapy. Neurology. 2017;89:1107–16.CrossRefPubMedPubMedCentral
36.
go back to reference Haghikia A, Dendrou CA, Schneider R, et al. Severe B-cell-mediated CNS disease secondary to alemtuzumab therapy. Lancet Neurol. 2017;16:104–6.CrossRefPubMed Haghikia A, Dendrou CA, Schneider R, et al. Severe B-cell-mediated CNS disease secondary to alemtuzumab therapy. Lancet Neurol. 2017;16:104–6.CrossRefPubMed
37.
go back to reference Ellebedy AH, Jackson KJ, Kissick HT, Nakaya HI, Davis CW, Roskin KM, McElroy AK, Oshansky CM, Elbein R, Thomas S, Lyon GM, Spiropoulou CF, Mehta AK, Thomas PG, Boyd SD, Ahmed R. Defining antigen-specific plasmablast and memory B cell subsets in human blood after viral infection or vaccination. Nat Immunol. 2016;17:1226–34.CrossRefPubMedPubMedCentral Ellebedy AH, Jackson KJ, Kissick HT, Nakaya HI, Davis CW, Roskin KM, McElroy AK, Oshansky CM, Elbein R, Thomas S, Lyon GM, Spiropoulou CF, Mehta AK, Thomas PG, Boyd SD, Ahmed R. Defining antigen-specific plasmablast and memory B cell subsets in human blood after viral infection or vaccination. Nat Immunol. 2016;17:1226–34.CrossRefPubMedPubMedCentral
38.
go back to reference Serafini B, Rosicarelli B, Magliozzi R, Stigliano E, Aloisi F. Detection of ectopic B-cell follicles with germinal centers in the meninges of patients with secondary progressive multiple sclerosis. Brain Pathol. 2004;14:164–74.CrossRefPubMed Serafini B, Rosicarelli B, Magliozzi R, Stigliano E, Aloisi F. Detection of ectopic B-cell follicles with germinal centers in the meninges of patients with secondary progressive multiple sclerosis. Brain Pathol. 2004;14:164–74.CrossRefPubMed
39.
go back to reference Magliozzi R, Howell O, Vora A, et al. Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain. 2007;130:1089–104.CrossRefPubMed Magliozzi R, Howell O, Vora A, et al. Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain. 2007;130:1089–104.CrossRefPubMed
40.
go back to reference Kalincik T, Brown JWL, Robertson N, et al. Treatment effectiveness of alemtuzumab compared with natalizumab, fingolimod, and interferon beta in relapsing-remitting multiple sclerosis: a cohort study. Lancet Neurol. 2017;16:271–81.CrossRefPubMed Kalincik T, Brown JWL, Robertson N, et al. Treatment effectiveness of alemtuzumab compared with natalizumab, fingolimod, and interferon beta in relapsing-remitting multiple sclerosis: a cohort study. Lancet Neurol. 2017;16:271–81.CrossRefPubMed
41.
go back to reference Harrer A, Tumani H, Niendorf S, et al. Cerebrospinal fluid parameters of B cell-related activity in patients with active disease during natalizumab therapy. Mult Scler. 2013;19:1209–12.CrossRefPubMed Harrer A, Tumani H, Niendorf S, et al. Cerebrospinal fluid parameters of B cell-related activity in patients with active disease during natalizumab therapy. Mult Scler. 2013;19:1209–12.CrossRefPubMed
42.
go back to reference von Glehn F, Farias AS, de Oliveira AC, et al. Disappearance of cerebrospinal fluid oligoclonal bands after natalizumab treatment of multiple sclerosis patients. Mult Scler. 2012;18:1038–41.CrossRefPubMed von Glehn F, Farias AS, de Oliveira AC, et al. Disappearance of cerebrospinal fluid oligoclonal bands after natalizumab treatment of multiple sclerosis patients. Mult Scler. 2012;18:1038–41.CrossRefPubMed
43.
go back to reference Bail K, Notz Q, Rovituso DM, et al. Differential effects of FTY720 on the B cell compartment in a mouse model of multiple sclerosis. J Neuroinflammation. 2017;14:148.CrossRefPubMedPubMedCentral Bail K, Notz Q, Rovituso DM, et al. Differential effects of FTY720 on the B cell compartment in a mouse model of multiple sclerosis. J Neuroinflammation. 2017;14:148.CrossRefPubMedPubMedCentral
44.
go back to reference Lowenstein H, Shah A, Chant A, Khan A. Different mechanisms of Campath-1H-mediated depletion for CD4 and CD8 T cells in peripheral blood. Transpl Int. 2006;19:927–36.CrossRefPubMed Lowenstein H, Shah A, Chant A, Khan A. Different mechanisms of Campath-1H-mediated depletion for CD4 and CD8 T cells in peripheral blood. Transpl Int. 2006;19:927–36.CrossRefPubMed
45.
go back to reference Komori M, Lin YC, Cortese I, et al. Insufficient disease inhibition by intrathecal rituximab in progressive multiple sclerosis. Ann Clin Transl Neurol. 2016;3:166–79.CrossRefPubMedPubMedCentral Komori M, Lin YC, Cortese I, et al. Insufficient disease inhibition by intrathecal rituximab in progressive multiple sclerosis. Ann Clin Transl Neurol. 2016;3:166–79.CrossRefPubMedPubMedCentral
46.
go back to reference Stys PK, Zamponi GW, van Minnen J, Geurts JJ. Will the real multiple sclerosis please stand up? Nat Rev Neurosci. 2012;13:507–14.CrossRefPubMed Stys PK, Zamponi GW, van Minnen J, Geurts JJ. Will the real multiple sclerosis please stand up? Nat Rev Neurosci. 2012;13:507–14.CrossRefPubMed
48.
go back to reference Jones JL, Anderson JM, Phuah CL, et al. Improvement in disability after alemtuzumab treatment of multiple sclerosis is associated with neuroprotective autoimmunity. Brain. 2010;133:2232–47.CrossRefPubMed Jones JL, Anderson JM, Phuah CL, et al. Improvement in disability after alemtuzumab treatment of multiple sclerosis is associated with neuroprotective autoimmunity. Brain. 2010;133:2232–47.CrossRefPubMed
Metadata
Title
Anti-CD52 antibody treatment depletes B cell aggregates in the central nervous system in a mouse model of multiple sclerosis
Authors
Micha Simon
Rojda Ipek
György A. Homola
Damiano M. Rovituso
Andrea Schampel
Christoph Kleinschnitz
Stefanie Kuerten
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2018
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-018-1263-9

Other articles of this Issue 1/2018

Journal of Neuroinflammation 1/2018 Go to the issue