Alternating Current Stimulation for Vision Restoration after Optic Nerve Damage: A Randomized Clinical Trial

Carolin Gall, Sein Schmidt, Michael P Schittkowski, Andrea Antal, Géza Gergely Ambrus, Walter Paulus, Moritz Dannhauer, Romualda Michalik, Alf Mante, Michal Bola, Anke Lux, Siegfried Kropf, Stephan A Brandt, Bernhard A Sabel, Carolin Gall, Sein Schmidt, Michael P Schittkowski, Andrea Antal, Géza Gergely Ambrus, Walter Paulus, Moritz Dannhauer, Romualda Michalik, Alf Mante, Michal Bola, Anke Lux, Siegfried Kropf, Stephan A Brandt, Bernhard A Sabel

Abstract

Background: Vision loss after optic neuropathy is considered irreversible. Here, repetitive transorbital alternating current stimulation (rtACS) was applied in partially blind patients with the goal of activating their residual vision.

Methods: We conducted a multicenter, prospective, randomized, double-blind, sham-controlled trial in an ambulatory setting with daily application of rtACS (n = 45) or sham-stimulation (n = 37) for 50 min for a duration of 10 week days. A volunteer sample of patients with optic nerve damage (mean age 59.1 yrs) was recruited. The primary outcome measure for efficacy was super-threshold visual fields with 48 hrs after the last treatment day and at 2-months follow-up. Secondary outcome measures were near-threshold visual fields, reaction time, visual acuity, and resting-state EEGs to assess changes in brain physiology.

Results: The rtACS-treated group had a mean improvement in visual field of 24.0% which was significantly greater than after sham-stimulation (2.5%). This improvement persisted for at least 2 months in terms of both within- and between-group comparisons. Secondary analyses revealed improvements of near-threshold visual fields in the central 5° and increased thresholds in static perimetry after rtACS and improved reaction times, but visual acuity did not change compared to shams. Visual field improvement induced by rtACS was associated with EEG power-spectra and coherence alterations in visual cortical networks which are interpreted as signs of neuromodulation. Current flow simulation indicates current in the frontal cortex, eye, and optic nerve and in the subcortical but not in the cortical regions.

Conclusion: rtACS treatment is a safe and effective means to partially restore vision after optic nerve damage probably by modulating brain plasticity. This class 1 evidence suggests that visual fields can be improved in a clinically meaningful way.

Trial registration: ClinicalTrials.gov NCT01280877.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Consort flow chart and study…
Fig 1. Consort flow chart and study design.
(A) Patient flow for cases included in the primary outcome measure analysis. Of 98 eligible patients, 45 were treated with rtACS and 37 with sham-stimulation. Five subjects left the study between initial screening and BASELINE for different reasons and another five subjects were excluded due to violation of an inclusion criterion (unacceptable fluctuations between initial screening and BASELINE). During the treatment phase three subjects dropped out because of medical conditions that were unrelated to study participation. Three treated cases of legally blind subjects were excluded from subsequent analyses due to violation of inclusion criterion (no residual vision). (B) Study design with diagnostic and treatment visits. Randomization was done after BASELINE assessment. Stability of VF defects was ascertained by comparing VFs at BASELINE with those obtained during the screening visit 2 weeks earlier. Upon completion of the 10-day treatment, all initial diagnostic tests were repeated (POST). The FOLLOW-UP diagnostic assessment was conducted after a therapy-free interval of at least 2 months.
Fig 2. Perimetry measures and EEG.
Fig 2. Perimetry measures and EEG.
(A) Primary and secondary analyses of VF outcome between- and within-groups after rtACS and sham-stimulation bar charts of primary (first upper graph) and secondary parameters of VF diagnostics measured using HRP and standard-automated static and kinetic perimetry. Results are given as medians and 95%-CI. Between-group comparisons were performed according to a pre-defined hypothesis using a one-sided U-test. Within-group BASELINE vs. POST and BASELINE vs. FOLLOW-UP comparisons were calculated separately for each treatment arm using Wilcoxon matched-pairs signed rank tests. The respective p-values are reported with p<0.05 considered as significant. (B) Individual change in HRP VF charts at BASELINE and POST in the two best responding patients of both groups. By superimposing HRP computer campimetric VF charts of three repeated measurements, VF areas were categorized as intact (perfect stimulus detection at a given location, white spots), partially damaged/relative defect (inconsistent stimulus detection, grey spots), and absolutely impaired areas (no stimulus detected, black spots). Detection increases and decreases after intervention are shown in blue and red, respectively. The percentage improvement of the detection accuracy was comparable between the whole HRP VF 16x21.5° and the central 5° VF. (C) Power spectra before and after the first stimulation session. Left sub-figure: One session of tACS increased power of theta (Z = 3.583, p<0.001), alpha (t = 4.571, p<0.001) and beta bands (Z = 3.142, p = 0.002) recorded from electrode positions above the visual cortex. Middle sub-figure: After sham stimulation a significant power increase was observed for only the theta band (Z = 3.147, p = 0.002). Right sub-figure: Scatter plot showing the relation between change in alpha band coherence at the occipital area of interest and change in detection accuracy in total visual field (primary outcome measure).
Fig 3
Fig 3
Visualization of simulated electrical fields during rtACS: current density maxima on eye/optical nerve (A), brain tissue surface (B) and in the volume (C). Although four electrodes were used for treatment, they were used only one at a time. Therefore, the current flow simulation was done with only one electrode, representing all other electrodes. (A) Current density maxima of about 0.0044 A/m2 can be observed on the upper part of the outer eye surface that is closest to the stimulating electrode. Furthermore, the optical nerve of the stimulated eye also receives parts of the stimulating current density magnitude as currents enter the inner skull. (B) Local current density maxima can be found at frontal brain regions spatially located close to the stimulating anodal electrode. (C) Another area of locally increased current density can be found at the brain stem and lower cerebellum.

References

    1. Ghaffarieh A, Levin LA. Optic nerve disease and axon pathophysiology. Int Rev Neurobiol 2012; 105: 1–17. 10.1016/B978-0-12-398309-1.00002-0
    1. Gall C, Lucklum J, Sabel BA, Franke GH. Vision- and health-related quality of life in patients with visual field loss after postchiasmatic lesions. Invest Ophthalmol Vis Sci 2009; 50: 2765–76. 10.1167/iovs.08-2519
    1. Greenwald D, Kapoor N, Singh AD. Visual impairments in the first year after traumatic brain injury. Brain Inj 2012; 26: 1338–59. 10.3109/02699052.2012.706356
    1. Raz N, Dotan S, Chokron S, Ben-Hur T, Levin N. Demyelination affects temporal aspects of perception: an optic neuritis study. Ann Neurol 2012; 71: 531–8. 10.1002/ana.22692
    1. Nair G, Renga V, Lindenberg R, Zhu L, Schlaug G. Optimizing recovery potential through simultaneous occupational therapy and non-invasive brain-stimulation using tDCS. Restor Neurol Neurosci 2011; 29: 411–20. 10.3233/RNN-2011-0612
    1. Chrysikou EG, Hamilton RH. Noninvasive brain stimulation in the treatment of aphasia: exploring interhemispheric relationships and their implications for neurorehabilitation. Restor Neurol Neurosci 2011; 29: 375–94. 10.3233/RNN-2011-0610
    1. Song S, Sandrini M, Cohen LG. Modifying somatosensory processing with non-invasive brain stimulation. Restor Neurol Neurosci 2011; 29: 427–37. 10.3233/RNN-2011-0614
    1. Antal A, Paulus W, Nitsche MA. Electrical stimulation and visual network plasticity. Restor Neurol Neurosci 2011; 29: 365–74. 10.3233/RNN-2011-0609
    1. Laczó B, Antal A, Niebergall R, Treue S, Paulus W. Transcranial alternating stimulation in a high gamma frequency range applied over V1 improves contrast perception but does not modulate spatial attention. Brain Stimul 2012; 5: 484–91. 10.1016/j.brs.2011.08.008
    1. Sabel BA, Henrich-Noack P, Fedorov A, Gall C. Vision restoration after brain and retina damage: the "residual vision activation theory". Prog Brain Res 2011; 192: 199–262. 10.1016/B978-0-444-53355-5.00013-0
    1. Zaehle T, Rach S, Herrmann CS. Transcranial alternating current stimulation enhances individual alpha activity in human EEG. PLoS One 2010; 5: 13766.
    1. Bola M, Gall C, Moewes C, Fedorov A, Hinrichs H, Sabel BA. Brain functional connectivity network breakdown and restoration in blindness. Neurology 2014; 83: 542–51. 10.1212/WNL.0000000000000672
    1. Connell N, Merabet LB. Uncovering the connectivity of the brain in relation to novel vision rehabilitation strategies. Neurology, 2014; 83:1–2.
    1. Sabel BA, Fedorov AB, Naue N, Borrmann A, Herrmann C, Gall C. Non-invasive alternating current stimulation improves vision in optic neuropathy. Restor Neurol Neurosci 2011; 29: 493–505. 10.3233/RNN-2011-0624
    1. Gall C, Sgorzaly S, Schmidt S, Brandt S, Fedorov A, Sabel BA. Noninvasive transorbital alternating current stimulation improves subjective visual functioning and vision-related quality of life in optic neuropathy. Brain Stimul 2011; 4: 175–88. 10.1016/j.brs.2011.07.003
    1. Schmidt S, Mante A, Rönnefarth M, Fleischmann R, Gall C, Brandt SA. Progressive enhancement of alpha activity and visual function in patients with optic neuropathy: a two-week repeated session alternating current stimulation study. Brain Stimul 2013; 6: 87–93. 10.1016/j.brs.2012.03.008
    1. Kasten E, Wüst S, Behrens-Baumann W, Sabel BA. Computer-based training for the treatment of partial blindness. Nat Med 1998; 4: 1083–7.
    1. Mangione M, Lee PP, Pitts J, Gutierrez P, Berry S, Hays RD. Psychometric properties of the National Eye Institute-Visual Function Questionnaire (NEI-VFQ): NEI-VFQ Field Test Investigators. Arch Ophthalmol 1998; 116: 1496–1504.
    1. Pereda E, Quiroga RQ, Bhattacharya J. Nonlinear multivariate analysis of neurophysiological signals. Prog Neurobiol 2005; 77: 1–37.
    1. Haueisen J, Tuch DS, Ramon C, Schimpf PH, Wedeen VJ, George JS, et al. The influence of brain tissue anisotropy on human EEG and MEG. Neuroimage 2002; 15: 159–66.
    1. Dannhauer M, Lanfer B, Wolters CH, Knösche TR. Modeling of the human skull in EEG source analysis. Hum Brain Mapp 2011; 32: 1383–99. 10.1002/hbm.21114
    1. Eichelbaum S, Dannhauer M, Hlawitschka M, Brooks D, Knösche TR, Scheuermann G. Visualization simulated electrical fields from electroencephalography and transcranial electric brain stimulation: a comparative evaluation. NeuroImage 2014; 101: 513–30. 10.1016/j.neuroimage.2014.04.085
    1. Lindenblatt G, Silny J. A model of the electrical volume conductor in the region of the eye in the ELF range. Phys Med Biol 2001; 46: 3051–9.
    1. Gall C, Steger B, Koehler J, Sabel BA. Evaluation of two treatment outcome prediction models for restoration of visual fields in patients with postchiasmatic visual pathway lesions. Neuropsychologia 2013; 51: 2271–80. 10.1016/j.neuropsychologia.2013.06.028
    1. Foik AT, Kublik E, Sergeeva EG, Tatlisumak T, Rossini PM, Sabel BA, et al. Retinal origin of electrically evoked potentials in response to transcorneal alternating current stimulation in the rat. Invest Ophthalmol Vis Sci 2015; 56: 1711–18. 10.1167/iovs.14-15617
    1. Poggel DA, Kasten E, Sabel BA. Attentional cueing improves vision restoration therapy in patients with visual field defects. Neurology 2004; 63: 2069–76.
    1. Sahraie A, Trevethan CT, Macleod MJ, Weiskrantz L, Hunt AR. The continuum of detection and awareness of visual stimuli within the blindfield: from blindsight to the sighted-sight. Invest Ophthalmol Vis Sci 2013; 54: 3579–85. 10.1167/iovs.12-11231
    1. Sabel BA, Gudlin J. Vision restoration training in glaucoma—A prospective, double-blind, randomized, placebo-controlled clinical trial. JAMA Ophthalmology 2014; 132: 381–9. 10.1001/jamaophthalmol.2013.7963
    1. Bola M, Gall C, Sabel BA. Disturbed temporal dynamics of brain synchronization in vision loss. Cortex, 2015a; DOI information: 10.1016/j.cortex.2015.03.020
    1. Bola M, Sabel BA. Dynamic reorganization of brain functional networks during cognition. Neuroimage 2015; 114: 398–413. 10.1016/j.neuroimage.2015.03.057
    1. Ali M, Sellers K, Frohlich F. Transcranial Alternating Current Stimulation Modulates Large-Scale Cortical Network Activity by Network Resonance. J Neurosci 2013; 33: 11262–75. 10.1523/JNEUROSCI.5867-12.2013
    1. Merlet I, Birot G, Salvador R, Molaee-Ardekani B, Mekonnen A, Soria-Frish A, et al. From oscillatory transcranial current stimulation to scalp EEG changes: a biophysical and physiological modeling study. PLoS One 2013; 8: e57330 10.1371/journal.pone.0057330
    1. Reato D, Rahman A, Bikson M, Parra L. Low-Intensity Electrical Stimulation Affects Network Dynamics by Modulating Population Rate and Spike Timing. J Neurosci 2010; 30: 15067–79. 10.1523/JNEUROSCI.2059-10.2010
    1. Schmidt SL, Iyengar AK, Foulser AA, Boyle MR, Fröhlich F. Endogenous cortical oscillations constrain neuromodulation by weak electric fields. Brain Stimul 2014; 7(6): 878–89. 10.1016/j.brs.2014.07.033
    1. Vosskuhl J, Huster R, Herrmann C. Increase in short-term memory capacity induced by down-regulating individual theta frequency via transcranial alternating current stimulation. Front Hum Neurosci 2015; 9: 257 10.3389/fnhum.2015.00257
    1. Schmidt S, Scholz M, Obermayer K, Brandt SA. Patterned brain stimulation. What a framework with rhythmic and noisy components might tell us about recovery maximization. Front Hum Neurosci 2013a; 7: 325.
    1. Schatz A, Röck T, Naycheva L, Willmann G, Wilhelm B, Peters T, et al. Transcorneal electrical stimulation for patients with retinitis pigmentosa: a prospective, randomized, sham-controlled exploratory study. Invest Ophthalmol Vis Sci 2011; 52: 4485–96. 10.1167/iovs.10-6932
    1. Plow B, Obretenova SN, Fregni F, Pascual-Leone A, Merabet LB. Comparison of VF training for hemianopia with active versus sham transcranial direct cortical stimulation. Neurorehabil Neural Repair 2012; 26: 616–26. 10.1177/1545968311431963
    1. Gall C, Silvennoinen K, Granata G, de Rossi F, Vecchio F, Brösel D, et al. Non-invasive electric current stimulation for restoration of vision after unilateral occipital stroke. Contemp Clin Trials 2015; 43: 231–6. 10.1016/j.cct.2015.06.005

Source: PubMed

Подписаться