Assessment of Opicinumab in Acute Optic Neuritis Using Multifocal Visual Evoked Potential

Alexander Klistorner, Yi Chai, Letizia Leocani, Philipp Albrecht, Orhan Aktas, Helmut Butzkueven, Tjalf Ziemssen, Focke Ziemssen, Jette Frederiksen, Lei Xu, Diego Cadavid, RENEW MF-VEP Investigators, Helmut Butzkueven, Ray Garrick, Ludo Vanopdenbosch, Jette Frederiksen, Orhan Aktas, Philipp Albrecht, Focke Ziemssen, Tjalf Ziemssen, Giancarlo Comi, Bernardo Sánchez Dalmau, Magnus Andersson, Gordon T Plant, Tim Matthews, Graeme Williams, Alexander Klistorner, Yi Chai, Letizia Leocani, Philipp Albrecht, Orhan Aktas, Helmut Butzkueven, Tjalf Ziemssen, Focke Ziemssen, Jette Frederiksen, Lei Xu, Diego Cadavid, RENEW MF-VEP Investigators, Helmut Butzkueven, Ray Garrick, Ludo Vanopdenbosch, Jette Frederiksen, Orhan Aktas, Philipp Albrecht, Focke Ziemssen, Tjalf Ziemssen, Giancarlo Comi, Bernardo Sánchez Dalmau, Magnus Andersson, Gordon T Plant, Tim Matthews, Graeme Williams

Abstract

Background: Multifocal visual evoked potential (MF-VEP) assesses a wider visual field than full-field VEP (FF-VEP) and potentially offers a more precise analysis of optic nerve injury and repair following optic neuritis. MF-VEP may offer advantages over FF-VEP as an endpoint in clinical trials of remyelinating therapies.

Objective: MF-VEP testing was used to study changes in visual pathways in 48% of RENEW [phase II, opicinumab (anti-LINGO-1; BIIB033) vs. placebo after first acute unilateral optic neuritis] participants.

Methods: This exploratory MF-VEP RENEW substudy compared mean outcomes at weeks 24 and 32 among participants in the intent-to-treat (ITT; n = 39; 72% female; mean age: 32.3 years) and per-protocol (PP; n = 31; 71% female; mean age: 32.2 years) populations in affected and fellow eye latency from fellow eye baseline latency and affected and fellow eye amplitude from their own baselines. Treatment differences were evaluated using analysis of covariance (week 24) and a mixed-effect model of repeated measures (week 32). Last observation carried forward was used to impute missing data at week 24.

Results: A trend for improvement in affected eye MF-VEP latency with opicinumab versus placebo was seen in the ITT and PP populations at weeks 24 and 32. Both treatment groups in the ITT population experienced partial recovery of amplitude in the affected eye at week 32. Notably, the mean change in fellow eye amplitude at weeks 24 and 32 was - 17.57 and - 31.41 nanovolts (nV) in placebo but only - 0.59 and 1.93 nV in the opicinumab group [differences at weeks 24 and 32: 16.98 nV (p = 0.050) and 33.33 nV (p < 0.01), respectively].

Conclusion: Results from this substudy showed advantages of MF-VEP over FF-VEP in multicenter studies of central nervous system reparative therapies and provide novel evidence that fellow eye visual pathway amplitude loss occurs after optic neuritis but can potentially be prevented by opicinumab treatment.

Registration: ClinicalTrials.gov identifier NCT01721161.

Conflict of interest statement

Alexander Klistorner has nothing to disclose. Yi Chai was a full-time employee of Biogen during the performance of the clinical trial, data analysis, and initial drafting of the manuscript. Letizia Leocani has received honoraria for consulting and/or speaking activities from Biogen, Merck, Roche, and Teva; research support from Biogen, Merck, and Novartis; and travel support from Almirall, Biogen, Merck, Novartis, and Roche. Philipp Albrecht has received grants from Biogen, the Dr. Robert Pfleger Foundation, Ipsen, Merz, and Novartis; received non-financial support from Bayer HealthCare, Ipsen, Merck, Merz, Novartis, Roche, and Teva; participated in advisory boards for Allergan, Ipsen, and Merz; and received personal fees from Allergan, Bayer HealthCare, Biogen, Ipsen, Novartis, Roche, and Teva, outside of the submitted work. Orhan Aktas has received advisor fees or honoraria from Almirall, Bayer HealthCare, Biogen, MedImmune, Merck, Novartis, and Teva; and research support from Bayer HealthCare, Biogen, Novartis, Roche, and Teva. Helmut Butzkueven has participated in advisory boards for Biogen, Merck, Novartis, and Teva; received consulting fees from Novartis and Oxford Pharmagenesis; lecture fees from Biogen; received payment for development of educational presentations from Biogen, Merck, and Novartis; and research grants from Biogen and Novartis, outside of the submitted work. Tjalf Ziemssen has received consulting fees from Almirall, Bayer HealthCare, Biogen, Genzyme, GlaxoSmithKline, Merck Serono, MSD, Novartis, Sanofi-Aventis, Synthon, and Teva; and research support from Bayer HealthCare, Biogen, Merck Serono, Novartis, Sanofi-Aventis, and Teva. Focke Ziemssen has received consulting fees from Alimera, Allergan, Bayer HealthCare, Boehringer Ingelheim, Novartis, and Roche; and research support from Novartis. Jette Frederiksen has participated in scientific advisory boards for Almirall, Biogen, Genzyme, Merck Serono, Novartis, Sanofi-Aventis, and Teva; received speaker honoraria from Biogen, Merck Serono, Santhera, and Teva; and been an advisor on preclinical development for Takeda, outside of the submitted work. Lei Xu was a full-time employee of Biogen during the performance of the clinical trial, data analysis, and initial drafting of the manuscript. Diego Cadavid was a full-time employee of Biogen during the performance of the clinical trial and a full-time employee of Fulcrum Therapeutics during submission of the manuscript; the work is not related to his employment in Fulcrum Therapeutics. He has patent WO 2016112270 A1: Lingo-1 antagonists and uses for treatment of demyelinating disorders, which is assigned to Biogen.

Figures

Fig. 1
Fig. 1
Individual segments assessed using a multifocal visual evoked potential and b reversing black-and-white checks used for monocular stimulation
Fig. 2
Fig. 2
Mean change in MF-VEP latency, adjusted for the baseline latency of unaffected fellow eye, at a week 24 and b week 32 in the affected eye compared with the unaffected fellow eye at baseline in the substudy ITT and PP populations. Error bars represent 95% CIs. aAnalyzed by analysis of covariance. bAnalyzed by mixed-effect model of repeated measures. CI confidence interval, ITT intent-to-treat, MF-VEP multifocal visual evoked potential, PP per protocol
Fig. 3
Fig. 3
Mean change in multifocal visual evoked potential amplitude by treatment from baseline over 32 weeks in the a affected eye and b unaffected fellow eye in the intent-to-treat population. In the heat map displays, green represents amplitude gains (improvement) and red represents amplitude losses (worsening) from baseline. Results for the per protocol population were not calculated. aCompared with baseline value of the affected eye. bCompared with baseline value of the fellow eye. CI confidence interval, nV nanovolts
Fig. 4
Fig. 4
Adjusted mean differences in a MF-VEP latency and b amplitude at week 24 in participants classified with latency recovery using the primary endpoint measure of FF-VEP. FF-VEP latency recovery was defined as affected eye FF-VEP latency at week 24 ≤ 10% worse than the baseline fellow eye; FF-VEP latency was the primary endpoint in RENEW. CI confidence interval, FF-VEP full-field visual evoked potential, MF-VEP multifocal visual evoked potential

References

    1. Mi S, Pepinsky RB, Cadavid D. Blocking LINGO-1 as a therapy to promote CNS repair: from concept to the clinic. CNS Drugs. 2013;27:493–503.
    1. Tran JQ, Rana J, Barkhof F, Melamed I, Gevorkyan H, Wattjes MP, et al. Randomized phase I trials of the safety/tolerability of anti-LINGO-1 monoclonal antibody BIIB033. Neurol Neuroimmunol Neuroinflamm. 2014;1:e18.
    1. Cadavid D, Balcer L, Galetta S, Aktas O, Ziemssen T, Vanopdenbosch L, RENEW Study Investigators et al. Safety and efficacy of opicinumab in acute optic neuritis (RENEW): a randomised, placebo-controlled, phase 2 trial. Lancet Neurol. 2017;16:189–199.
    1. Halliday AM, McDonald WI, Mushin J. Delayed visual evoked response in optic neuritis. Lancet. 1972;1:982–985.
    1. Hood DC, Odel JG, Winn BJ. The multifocal visual evoked potential. J Neuroophthalmol. 2003;23:279–289.
    1. Klistorner A, Arvind H, Garrick R, Graham SL, Paine M, Yiannikas C. Interrelationship of optical coherence tomography and multifocal visual-evoked potentials after optic neuritis. Investig Ophthalmol Vis Sci. 2010;51:2770–2777.
    1. Creel DJ. Visual evoked potentials. . Accessed 5 Jul 2016.
    1. Pihl-Jensen G, Schmidt MF, Frederiksen JL. Multifocal visual evoked potentials in optic neuritis and multiple sclerosis: a review. Clin Neurophysiol. 2017;128:1234–1245.
    1. Klistorner AI, Graham SL, Grigg JR, Billson FA. Multifocal topographic visual evoked potential: improving objective detection of local visual field defects. Investig Ophthalmol Vis Sci. 1998;39:937–950.
    1. Hood DC, Odel JG, Zhang X. Tracking the recovery of local optic nerve function after optic neuritis: a multifocal VEP study. Investig Ophthalmol Vis Sci. 2000;41:4032–4038.
    1. Narayanan D, Cheng H, Tang RA, Frishman LJ. Reproducibility of multifocal visual evoked potential and traditional visual evoked potential in normal and multiple sclerosis eyes. Doc Ophthalmol. 2015;130:31–41.
    1. Nebbioso M, Steigerwalt RD, Pecori-Giraldi J, Vingolo EM. Multifocal and pattern-reversal visual evoked potentials vs. automated perimetry frequency-doubling technology matrix in optic neuritis. Indian J Ophthalmol. 2013;61:59–64.
    1. Cadavid D, Levin N, Costello F, Rahilly A, Klistorner A. Technical feasibility of implementing multifocal VEP for multicenter clinical trials. Neurology. 2013;80(P02):245.
    1. Klistorner A, Graham S, Fraser C, Garrick R, Nguyen T, Paine M, et al. Electrophysiological evidence for heterogeneity of lesions in optic neuritis. Investig Ophthalmol Vis Sci. 2007;48:4549–4556.
    1. Klistorner A, Arvind H, Garrick R, Yiannikas C, Paine M, Graham SL. Remyelination of optic nerve lesions: spatial and temporal factors. Mult Scler. 2010;16:786–795.
    1. Jones SJ, Brusa A. Neurophysiological evidence for long-term repair of MS lesions: implications for axon protection. J Neurol Sci. 2003;206:193–198.
    1. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. ICH harmonized tripartite guideline: guideline for good clinical practice. . Accessed 5 Jul 2016.
    1. World Medical Association WMA Declaration of Helsinki. Ethical principles for medical research involving human subjects. JAMA. 2013;310:2191–2194.
    1. Cruz-Herranz A, Balk LJ, Oberwahrenbrock T, Saidha S, Martinez-Lapiscina EH, Lagreze WA, IMSVISUAL Consortium et al. The APOSTEL recommendations for reporting quantitative optical coherence tomography studies. Neurology. 2016;86:2303–2309.
    1. Alshowaeir D, Yiannikas C, Garrick R, Parratt J, Barnett MH, Graham SL, et al. Latency of multifocal visual evoked potentials in nonoptic neuritis eyes of multiple sclerosis patients associated with optic radiation lesions. Investig Ophthalmol Vis Sci. 2014;55:3758–3764.
    1. Klistorner AI, Graham SL. Electroencephalogram-based scaling of multifocal visual evoked potentials: effect on intersubject amplitude variability. Investig Ophthalmol Vis Sci. 2001;42:2145–2152.
    1. Hubel DH, Wiesel TN. Receptive fields, binocular interaction and functional architecture in the cat’s visual cortex. J Physiol. 1962;160:106–154.
    1. Klistorner A, Vootakuru N, Wang C, Yiannikas C, Graham SL, Parratt J, et al. Decoding diffusivity in multiple sclerosis: analysis of optic radiation lesional and non-lesional white matter. PLoS One. 2015;10:e0122114.
    1. Rosenblatt MA, Behrens MM, Zweifach PH, Forman S, Odel JG, Duncan CM, et al. Magnetic resonance imaging of optic tract involvement in multiple sclerosis. Am J Ophthalmol. 1987;104:74–79.
    1. Frederiksen JL, Larsson HB, Nordenbo AM, Seedorff HH. Plaques causing hemianopsia or quadrantanopsia in multiple sclerosis identified by MRI and VEP. Acta Ophthalmol (Copenh). 1991;69:169–177.
    1. Dasenbrock HH, Smith SA, Ozturk A, Farrell SK, Calabresi PA, Reich DS. Diffusion tensor imaging of the optic tracts in multiple sclerosis: association with retinal thinning and visual disability. J Neuroimaging. 2011;21:e41–e49.
    1. Balcer LJ, Miller DH, Reingold SC, Cohen JA. Vision and vision-related outcome measures in multiple sclerosis. Brain. 2015;138:11–27.
    1. Gresle MM, Liu Y, Kilpatrick TJ, Kemper D, Wu QZ, Hu B, et al. Blocking LINGO-1 in vivo reduces degeneration and enhances regeneration of the optic nerve. Mult Scler J Exp Transl Clin. 2016;2:2055217316641704.
    1. Ranger A, Ray S, Szak S, Dearth A, Allaire N, Murray R, et al. Anti-LINGO-1 has no detectable immunomodulatory effects in preclinical and phase 1 studies. Neurol Neuroimmunol Neuroinflamm. 2017;5:e417.
    1. Prins D, Hanekamp S, Cornelissen FW. Structural brain MRI studies in eye diseases: are they clinically relevant? A review of current findings. Acta Ophthalmol. 2016;94:113–121.
    1. Balk LJ, Twisk JW, Steenwijk MD, Daams M, Tewarie P, Killestein J, et al. A dam for retrograde axonal degeneration in multiple sclerosis? J Neurol Neurosurg Psychiatry. 2014;85:782–789.
    1. Mi S, Hu B, Hahm K, Luo Y, Kam Hui ES, Yuan Q, et al. LINGO-1 antagonist promotes spinal cord remyelination and axonal integrity in MOG-induced experimental autoimmune encephalomyelitis. Nat Med. 2007;13:1228–1233.
    1. Mi S, Miller RH, Lee X, Scott ML, Shulag-Morskaya S, Shao Z, et al. LINGO-1 negatively regulates myelination by oligodendrocytes. Nat Neurosci. 2005;8:745–751.
    1. Lee X, Yang Z, Shao Z, Rosenberg SS, Levesque M, Pepinsky RB, et al. NGF regulates the expression of axonal LINGO-1 to inhibit oligodendrocyte differentiation and myelination. J Neurosci. 2007;27:220–225.
    1. Mi S, Miller RH, Tang W, Lee X, Hu B, Wu W, et al. Promotion of central nervous system remyelination by induced differentiation of oligodendrocyte precursor cells. Ann Neurol. 2009;65:304–315.
    1. Hu B, So KF, Pepinsky B, Mi S. Blocking LINGO-1 promotes axonal regeneration in the rat optic nerve crush model. Mult Scler. 2013;19:139.
    1. Martins Rosa A, Silva MF, Ferreira S, Murta J, Castelo-Branco M. Plasticity in the human visual cortex: an ophthalmology-based perspective. Biomed Res Int. 2013;2013:568354.
    1. Werring DJ, Bullmore ET, Toosy AT, Miller DH, Barker GJ, MacManus DG, et al. Recovery from optic neuritis is associated with a change in the distribution of cerebral response to visual stimulation: a functional magnetic resonance imaging study. J Neurol Neurosurg Psychiatry. 2000;68:441–449.
    1. Hickman SJ, Toosy AT, Jones SJ, Altmann DR, Miszkiel KA, MacManus DG, et al. Serial magnetization transfer imaging in acute optic neuritis. Brain. 2004;127:692–700.
    1. Wandell BA, Smirnakis SM. Plasticity and stability of visual field maps in adult primary visual cortex. Nat Rev Neurosci. 2009;10:873–884.
    1. Wong AM. New concepts concerning the neural mechanisms of amblyopia and their clinical implications. Can J Ophthalmol. 2012;47:399–409.
    1. Iobbi C, Korte M, Zagrebelsky M. Nogo-66 restricts synaptic strengthening via Lingo1 and the ROCK2-Cofilin pathway to control actin dynamics. Cereb Cortex. 2017;27:2779–2792.
    1. Kellner Y, Fricke S, Kramer S, Iobbi C, Wierenga CJ, Schwab ME, et al. Nogo-A controls structural plasticity at dendritic spines by rapidly modulating actin dynamics. Hippocampus. 2016;26:816–831.
    1. Kuo SH, Tang G, Louis ED, Ma K, Babji R, Balatbat M, et al. Lingo-1 expression is increased in essential tremor cerebellum and is present in the basket cell pinceau. Acta Neuropathol. 2013;125:879–889.

Source: PubMed

Подписаться