Immunological and short-term brain volume changes in relapsing forms of multiple sclerosis treated with interferon beta-1a subcutaneously three times weekly: an open-label two-arm trial

Michael G Dwyer, Robert Zivadinov, Yazhong Tao, Xin Zhang, Cheryl Kennedy, Niels Bergsland, Deepa P Ramasamy, Jackie Durfee, David Hojnacki, Bianca Weinstock-Guttman, Brooke Hayward, Fernando Dangond, Silva Markovic-Plese, Michael G Dwyer, Robert Zivadinov, Yazhong Tao, Xin Zhang, Cheryl Kennedy, Niels Bergsland, Deepa P Ramasamy, Jackie Durfee, David Hojnacki, Bianca Weinstock-Guttman, Brooke Hayward, Fernando Dangond, Silva Markovic-Plese

Abstract

Background: Brain volume atrophy is observed in relapsing-remitting multiple sclerosis (RRMS).

Methods: Brain volume changes were evaluated in 23 patients with RRMS treated with interferon β-1a 44 μg given subcutaneously (SC) three times a week (tiw) and 15 healthy controls. Percentages of whole brain and tissue-specific volume change were measured from baseline (0 months) to 3 months, from 3 to 6 months, and from baseline to 6 months using SIENAX Multi Time Point (SX-MTP) algorithms. Immunological status of patients was also determined and correlations between subsets of T cells and changes in brain volume were assessed.

Results: Interferon β-1a 44 μg SC tiw in 23 patients with RRMS resulted in significant reductions in whole brain and gray matter tissue volume early in the treatment course (baseline to 3 months; mean change; -0.95%; P = 0.030, -1.52%; P = 0.004, respectively), suggesting a short-term treatment-induced pseudoatrophy effect. From baseline to 6 months, there were significant correlations observed between decreased T- cell expression of IL-17 F and decreased whole brain and brain tissue-specific volume.

Conclusions: These findings are consistent with the interpretation of the pseudoatrophy effect as resolution of inflammation following treatment initiation with interferon β-1a 44 μg SC tiw, rather than disease-related tissue loss.

Trial registration: ClinicalTrials.gov; NCT01085318.

Figures

Fig. 1
Fig. 1
Brain volume changes in RRMS patients vs HCs. Percent change in whole brain volume in RRMS patients treated with IFN β-1a SC tiw and in HCs was measured. No P-values remained significant after adjustment for multiple comparisons. *P < 0.05, within-group difference from zero (Wilcoxon signed-rank test). †P < 0.05, patients versus HCs (Wilcoxon rank-sum test). In the box plots, the bold line represents the median; the boxes represent the middle 50 % of data; the top and bottom of the box represent the third and first quartiles; the open circles are outliers. The whisker lines above and below the boxes represent the largest and smallest values that are not considered to be outliers. Means are denoted by a ‘+’ sign. HC healthy control; IFN interferon; RRMS relapsing–remitting multiple sclerosis; SC subcutaneously; tiw three times weekly
Fig. 2
Fig. 2
Gray matter volume changes in RRMS patients and HCs. Percent change in GM volume in RRMS patients treated with IFN β-1a SC tiw and in HCs was assessed. P-values were for the difference from zero within groups from the Wilcoxon signed-rank test. P-values in parentheses are those remaining significant after Holm–Bonferroni correction for multiple comparisons. *P < 0.05. **P < 0.01. ***P < 0.001. In the box plots, the bold line represents the median; the boxes represent the middle 50 % of data; the top and bottom of the box represent the third and first quartiles; the open circles are outliers. The whisker lines above and below the boxes represent the largest and smallest values that are not considered to be outliers. Means are denoted by a ‘+’ sign. GM gray matter; HC healthy control; IFN interferon; RRMS relapsing–remitting multiple sclerosis; SC subcutaneously; tiw three times weekly
Fig. 3
Fig. 3
White matter volume changes in RRMS and HCs. Percent change in WM volume in RRMS patients treated with IFN β-1a SC tiw and in HCs was measured. Changes in WM volume were not significant at any timepoint. In the box plots, the bold line represents the median; the boxes represent the middle 50 % of data; the top and bottom of the box represent the third and first quartiles; the open circles are outliers. The whisker lines above and below the boxes represent the largest and smallest values that are not considered to be outliers. Means are denoted by a ‘+’ sign. HC healthy control; IFN interferon; RRMS relapsing–remitting multiple sclerosis; SC subcutaneously; tiw three times weekly; WM white matter
Fig. 4
Fig. 4
IL-17 F–expressing CD4+ T cells and brain volume changes in RRMS patients and HCs. Decreased percentage of IL-17 F–expressing CD4+ T cells from baseline to 6 months correlated with decreased whole brain volume from baseline to 6 months in RRMS patients treated with IFN β-1a SC tiw. HC healthy control; IFN interferon; IL interleukin; RRMS relapsing–remitting multiple sclerosis; SC subcutaneously; tiw three times weekly
Fig. 5
Fig. 5
IL-17 F–expressing CD8+ T cells and GM volume changes in RRMS. Higher percentage of IL-17 F–expressing CD8+ T cells from baseline to 6 months correlated with smaller decreases in GM volume from baseline to 6 months in treated patients with RRMS. GM gray matter; IL interleukin; RRMS relapsing–remitting multiple sclerosis
Fig. 6
Fig. 6
IL-17 F-expressing CD4+ T cells and WM volume changes in RRMS. Decreased percentage of IL-17 F-expressing CD4+ T cells from baseline to 6 months correlated with decreased WM volume from baseline to 6 months in treated patients with RRMS. IL interleukin; RRMS relapsing–remitting multiple sclerosis; WM white matter
Fig. 7
Fig. 7
IL-22–expressing CD4+ T cells and GM volume changes in RRMS. Higher percentage of IL-22–expressing CD4+ T cells at baseline correlated with smaller decreases in GM volume from baseline to 6 months in treated patients with RRMS. GM gray matter; IL interleukin; RRMS relapsing–remitting multiple sclerosis

References

    1. Rudick RA, Fisher E, Lee JC, Simon J, Jacobs L. Use of the brain parenchymal fraction to measure whole brain atrophy in relapsing-remitting MS. Multiple Sclerosis Collaborative Research Group. Neurology. 1999;53:1698–704. doi: 10.1212/WNL.53.8.1698.
    1. Calabrese M, Agosta F, Rinaldi F, Mattisi I, Grossi P, Favaretto A, et al. Cortical lesions and atrophy associated with cognitive impairment in relapsing-remitting multiple sclerosis. Arch Neurol. 2009;66:1144–50. doi: 10.1001/archneurol.2009.174.
    1. Luks TL, Goodkin TE, Nelson SJ, Majumdar S, Bacchetti P, Portnoy D, et al. A longitudinal study of ventricular volume in early relapsing-remitting multiple sclerosis. Mult Scler. 2000;6:332–7. doi: 10.1177/135245850000600507.
    1. Simon JH, Lull J, Jacobs LD, Rudick RA, Cookfair DL, Herndon RM, et al. A longitudinal study of T1 hypointense lesions in relapsing MS: MSCRG trial of interferon beta-1a. Neurology. 2000;55:185–92. doi: 10.1212/WNL.55.2.185.
    1. Frank JA, Richert N, Bash C, Stone L, Calabresi PA, Lewis B, et al. Interferon-beta-1b slows progression of atrophy in RRMS: Three-year follow-up in NAb- and NAb + patients. Neurology. 2004;62:719–25. doi: 10.1212/01.WNL.0000113765.75855.19.
    1. Ge Y, Grossman RI, Udupa JK, Fulton J, Constantinescu CS, Gonzales-Scarano F, et al. Glatiramer acetate (Copaxone) treatment in relapsing-remitting MS: quantitative MR assessment. Neurology. 2000;54:813–7. doi: 10.1212/WNL.54.4.813.
    1. Rinaldi F, Calabrese M, Seppi D, Puthenparampil M, Perini P, Gallo P. Natalizumab strongly suppresses cortical pathology in relapsing-remitting multiple sclerosis. Mult Scler. 2012;18:1760–7. doi: 10.1177/1352458512447704.
    1. van den Elskamp I, Lembcke J, Dattola V, Beckmann K, Pohl C, Hong W, et al. Persistent T1 hypointensity as an MRI marker for treatment efficacy in multiple sclerosis. Mult Scler. 2008;14:764–9. doi: 10.1177/1352458507087842.
    1. Vidal-Jordana A, Sastre-Garriga J, Perez-Miralles F, Tur C, Tintore M, Horga A, et al. Early brain pseudoatrophy while on natalizumab therapy is due to white matter volume changes. Mult Scler. 2013;19:1175–81. doi: 10.1177/1352458512473190.
    1. Wolinsky JS, Narayana PA, Johnson KP. United States open-label glatiramer acetate extension trial for relapsing multiple sclerosis: MRI and clinical correlates. Multiple Sclerosis Study Group and the MRI Analysis Center. Mult Scler. 2001;7:33–41. doi: 10.1177/135245850100700107.
    1. Jones CK, Riddehough A, Li DKB, Zhao GJ, Paty DW. MRI cerebral atrophy in relapsing remitting MS: results of the PRISMS trial. Neurology. 2001;56:A379.
    1. Paolillo A, Pozzilli C, Giugni E, Tomassini V, Gasperini C, Fiorelli M, et al. A 6-year clinical and MRI follow-up study of patients with relapsing-remitting multiple sclerosis treated with interferon-beta. Eur J Neurol. 2002;9:645–55. doi: 10.1046/j.1468-1331.2002.00476.x.
    1. PRISMS Study Group, University of British Columbia MS/MRI Analysis Group PRISMS-4: long-term efficacy of interferon-beta-1a in relapsing MS. Neurology. 2001;56:1628–36. doi: 10.1212/WNL.56.12.1628.
    1. Rovaris M, Comi G, Rocca MA, Wolinsky JS, Filippi M. Short-term brain volume change in relapsing-remitting multiple sclerosis: effect of glatiramer acetate and implications. Brain. 2001;124:1803–12. doi: 10.1093/brain/124.9.1803.
    1. Sormani MP, Rovaris M, Valsasina P, Wolinsky JS, Comi G, Filippi M. Measurement error of two different techniques for brain atrophy assessment in multiple sclerosis. Neurology. 2004;62:1432–4. doi: 10.1212/01.WNL.0000120663.85143.B3.
    1. Turner B, Lin X, Calmon G, Roberts N, Blumhardt LD. Cerebral atrophy and disability in relapsing-remitting and secondary progressive multiple sclerosis over four years. Mult Scler. 2003;9:21–7. doi: 10.1191/1352458503ms868oa.
    1. Gasperini C, Paolillo A, Giugni E, Galgani S, Bagnato F, Mainero C, et al. MRI brain volume changes in relapsing-remitting multiple sclerosis patients treated with interferon beta-1a. Mult Scler. 2002;8:119–23. doi: 10.1191/1352458502ms788oa.
    1. Khan O, Bao F, Shah M, Caon C, Tselis A, Bailey R, et al. Effect of disease-modifying therapies on brain volume in relapsing-remitting multiple sclerosis: results of a five-year brain MRI study. J Neurol Sci. 2012;312:7–12. doi: 10.1016/j.jns.2011.08.034.
    1. Zivadinov R, Reder AT, Filippi M, Minagar A, Stuve O, Lassmann H, et al. Mechanisms of action of disease-modifying agents and brain volume changes in multiple sclerosis. Neurology. 2008;71:136–44. doi: 10.1212/01.wnl.0000316810.01120.05.
    1. Riley C, Azevedo C, Bailey M, Pelletier D. Clinical applications of imaging disease burden in multiple sclerosis: MRI and advanced imaging techniques. Expert Rev Neurother. 2012;12:323–33. doi: 10.1586/ern.11.196.
    1. Filippi M, Rocca MA. Preventing brain atrophy should be the gold standard of effective theraphy in MS (after the first year of treatment): No. Mult Scler. 2013;19:1005–6. doi: 10.1177/1352458513482387.
    1. Ramgolam VS, Sha Y, Jin J, Zhang X, Markovic-Plese S. IFN-beta inhibits human Th17 cell differentiation. J Immunol. 2009;183:5418–27. doi: 10.4049/jimmunol.0803227.
    1. Battistini L, Piccio L, Rossi B, Bach S, Galgani S, Gasperini C, et al. CD8+ T cells from patients with acute multiple sclerosis display selective increase of adhesiveness in brain venules: a critical role for P-selectin glycoprotein ligand-1. Blood. 2003;101:4775–82. doi: 10.1182/blood-2002-10-3309.
    1. Babbe H, Roers A, Waisman A, Lassmann H, Goebels N, Hohlfeld R, et al. Clonal expansions of CD8(+) T cells dominate the T cell infiltrate in active multiple sclerosis lesions as shown by micromanipulation and single cell polymerase chain reaction. J Exp Med. 2000;192:393–404. doi: 10.1084/jem.192.3.393.
    1. Mars LT, Saikali P, Liblau RS, Arbour N. Contribution of CD8 T lymphocytes to the immuno-pathogenesis of multiple sclerosis and its animal models. Biochim Biophys Acta. 1812;2011:151–61.
    1. Li DK, Paty DW. UBC MS/MRI Analysis Research Group, PRISMS Study Group. Magnetic resonance imaging results of the PRISMS trial: a randomized, double-blind, placebo-controlled study of interferon-beta1a in relapsing-remitting multiple sclerosis. Ann Neurol. 1999;46:197–206. doi: 10.1002/1531-8249(199908)46:2<197::AID-ANA9>;2-P.
    1. Panitch H, Goodin D, Francis G, Chang P, Coyle P, O'Connor P, et al. Benefits of high-dose, high-frequency interferon beta-1a in relapsing-remitting multiple sclerosis are sustained to 16 months: final comparative results of the EVIDENCE trial. J Neurol Sci. 2005;239:67–74. doi: 10.1016/j.jns.2005.08.003.
    1. PRISMS Study Group Randomised double-blind placebo-controlled study of interferon beta-1a in relapsing/remitting multiple sclerosis. Lancet. 1998;352:1498–504. doi: 10.1016/S0140-6736(98)03334-0.
    1. Dwyer MG, Bergsland N, Zivadinov R. Improved longitudinal gray and white matter atrophy assessment via application of a 4-dimensional hidden Markov random field model. Neuroimage. 2014;90:207–17. doi: 10.1016/j.neuroimage.2013.12.004.
    1. Smith SM, Zhang Y, Jenkinson M, Chen J, Matthews PM, Federico A, et al. Accurate, robust, and automated longitudinal and cross-sectional brain change analysis. Neuroimage. 2002;17:479–89. doi: 10.1006/nimg.2002.1040.
    1. Zivadinov R, Dwyer MG, Markovic-Plese S, Kennedy C, Bergsland N, Ramasamy DP, et al. Effect of treatment with interferon beta-1a on changes in voxel-wise magnetization transfer ratio in normal appearing brain tissue and lesions of patients with relapsing-remitting multiple sclerosis: a 24-week, controlled pilot study. PLoS One. 2014;9:e91098. doi: 10.1371/journal.pone.0091098.
    1. Polman CH, Reingold SC, Banwell B, Clanet M, Cohen JA, Filippi M, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol. 2011;69:292–302. doi: 10.1002/ana.22366.
    1. Sled JG, Zijdenbos AP, Evans AC. A nonparametric method for automatic correction of intensity nonuniformity in MRI data. IEEE Trans Med Imaging. 1998;17:87–97. doi: 10.1109/42.668698.
    1. Nyul LG, Udupa JK, Zhang X. New variants of a method of MRI scale standardization. IEEE Trans Med Imaging. 2000;19:143–50. doi: 10.1109/42.836373.
    1. Kelemen A, Dwyer MG, Horakova D, Ticha V, Havrdova E, Zivadinov R. Measurement of gray matter volume is less susceptive to pseudoatrophy effect than that of white matter or whole brain volume in patients with multiple sclerosis. Mult Scler. 2008;14:S29–294. doi: 10.1177/1352458508096399.
    1. De SN, Airas L, Grigoriadis N, Mattle HP, O'Riordan J, Oreja-Guevara C, et al. Clinical relevance of brain volume measures in multiple sclerosis. CNS Drugs. 2014;28:147–56. doi: 10.1007/s40263-014-0140-z.
    1. Horakova D, Kalincik T, Dusankova JB, Dolezal O. Clinical correlates of grey matter pathology in multiple sclerosis. BMC Neurol. 2012;12:10. doi: 10.1186/1471-2377-12-10.
    1. Zivadinov R, Bagnato F, Nasuelli D, Bastianello S, Bratina A, Locatelli L, et al. Short-term brain atrophy changes in relapsing-remitting multiple sclerosis. J Neurol Sci. 2004;223:185–93. doi: 10.1016/j.jns.2004.05.010.
    1. Geurts JJ, Calabrese M, Fisher E, Rudick RA. Measurement and clinical effect of grey matter pathology in multiple sclerosis. Lancet Neurol. 2012;11:1082–92. doi: 10.1016/S1474-4422(12)70230-2.
    1. Calabrese M, Magliozzi R, Ciccarelli O, Geurts JJ, Reynolds R, Martin R. Exploring the origins of grey matter damage in multiple sclerosis. Nat Rev Neurosci. 2015;16:147–58. doi: 10.1038/nrn3900.
    1. Bo L, Vedeler CA, Nyland H, Trapp BD, Mork SJ. Intracortical multiple sclerosis lesions are not associated with increased lymphocyte infiltration. Mult Scler. 2003;9:323–31. doi: 10.1191/1352458503ms917oa.
    1. Peterson JW, Bo L, Mork S, Chang A, Trapp BD. Transected neurites, apoptotic neurons, and reduced inflammation in cortical multiple sclerosis lesions. Ann Neurol. 2001;50:389–400. doi: 10.1002/ana.1123.
    1. Lucchinetti CF, Popescu BF, Bunyan RF, Moll NM, Roemer SF, Lassmann H, et al. Inflammatory cortical demyelination in early multiple sclerosis. N Engl J Med. 2011;365:2188–97. doi: 10.1056/NEJMoa1100648.
    1. Popescu BF, Bunyan RF, Parisi JE, Ransohoff RM, Lucchinetti CF. A case of multiple sclerosis presenting with inflammatory cortical demyelination. Neurology. 2011;76:1705–10. doi: 10.1212/WNL.0b013e31821a44f1.
    1. Weinstock-Guttman B, Zivadinov R, Tamano-Blanco M, Abdelrahman N, Badgett D, Durfee J, et al. Immune cell BDNF secretion is associated with white matter volume in multiple sclerosis. J Neuroimmunol. 2007;188:167–74. doi: 10.1016/j.jneuroim.2007.06.003.
    1. Chard DT, Griffin CM, Parker GJ, Kapoor R, Thompson AJ, Miller DH. Brain atrophy in clinically early relapsing-remitting multiple sclerosis. Brain. 2002;125:327–37. doi: 10.1093/brain/awf025.
    1. Lim KO, Zipursky RB, Watts MC, Pfefferbaum A. Decreased gray matter in normal aging: an in vivo magnetic resonance study. J Gerontol. 1992;47:B26–30. doi: 10.1093/geronj/47.1.B26.
    1. Passe TJ, Rajagopalan P, Tupler LA, Byrum CE, MacFall JR, Krishnan KR. Age and sex effects on brain morphology. Prog Neuropsychopharmacol Biol Psychiatry. 1997;21:1231–7. doi: 10.1016/S0278-5846(97)00160-7.
    1. Pfefferbaum A, Mathalon DH, Sullivan EV, Rawles JM, Zipursky RB, Lim KO. A quantitative magnetic resonance imaging study of changes in brain morphology from infancy to late adulthood. Arch Neurol. 1994;51:874–87. doi: 10.1001/archneur.1994.00540210046012.

Source: PubMed

3
Subscribe