Development of a Sensitive Outcome for Economical Drug Screening for Progressive Multiple Sclerosis Treatment

Peter Kosa, Danish Ghazali, Makoto Tanigawa, Chris Barbour, Irene Cortese, William Kelley, Blake Snyder, Joan Ohayon, Kaylan Fenton, Tanya Lehky, Tianxia Wu, Mark Greenwood, Govind Nair, Bibiana Bielekova, Peter Kosa, Danish Ghazali, Makoto Tanigawa, Chris Barbour, Irene Cortese, William Kelley, Blake Snyder, Joan Ohayon, Kaylan Fenton, Tanya Lehky, Tianxia Wu, Mark Greenwood, Govind Nair, Bibiana Bielekova

Abstract

Therapeutic advance in progressive multiple sclerosis (MS) has been very slow. Based on the transformative role magnetic resonance imaging (MRI) contrast-enhancing lesions had on drug development for relapsing-remitting MS, we consider the lack of sensitive outcomes to be the greatest barrier for developing new treatments for progressive MS. The purpose of this study was to compare 58 prospectively acquired candidate outcomes in the real-world situation of progressive MS trials to select and validate the best-performing outcome. The 1-year pre-treatment period of adaptively designed IPPoMS (ClinicalTrials.gov #NCT00950248) and RIVITaLISe (ClinicalTrials.gov #NCT01212094) Phase II trials served to determine the primary outcome for the subsequent blinded treatment phase by comparing 8 clinical, 1 electrophysiological, 1 optical coherence tomography, 7 MRI volumetric, 9 quantitative T1 MRI, and 32 diffusion tensor imaging MRI outcomes. Fifteen outcomes demonstrated significant progression over 1 year (Δ) in the predetermined analysis and seven out of these were validated in two independent cohorts. Validated MRI outcomes had limited correlations with clinical scales, relatively poor signal-to-noise ratios (SNR) and recorded overlapping values between healthy subjects and MS patients with moderate-severe disability. Clinical measures correlated better, even though each reflects a somewhat different disability domain. Therefore, using machine-learning techniques, we developed a combinatorial weight-adjusted disability score (CombiWISE) that integrates four clinical scales: expanded disability status scale (EDSS), Scripps neurological rating scale, 25 foot walk and 9 hole peg test. CombiWISE outperformed all clinical scales (Δ = 9.10%; p = 0.0003) and all MRI outcomes. CombiWISE recorded no overlapping values between healthy subjects and disabled MS patients, had high SNR, and predicted changes in EDSS in a longitudinal assessment of 98 progressive MS patients and in a cross-sectional cohort of 303 untreated subjects. One point change in EDSS corresponds on average to 7.50 point change in CombiWISE with a standard error of 0.10. The novel validated clinical outcome, CombiWISE, outperforms the current broadly utilized MRI brain atrophy outcome and more than doubles sensitivity in detecting clinical deterioration in progressive MS in comparison to the scale traditionally used for regulatory approval, EDSS.

Keywords: clinical trial; composite scale; disability scale; multiple sclerosis; outcome measure; progressive MS; quantitative MRI.

Figures

Figure 1
Figure 1
Correlation matrix for 14 clinical and MRI measures in the cohort of 98 progressive MS patients. Correlation matrix for Mo −12 cross-sectional data (above the diagonal) and relative percentage change (Δ) over 1 year (below the diagonal) in the progressive MS cohort. The heatmap shows positive (shades of blue) and negative (shades of red) Spearman correlations. Black border of a window indicates Spearman correlation with p < 0.0018 (the Bonferroni-adjusted significance level for multiple comparisons of 28 tested variables). For exact values of Spearman correlation coefficients, p-values, and number observations, see Table S2 in Supplementary Material.
Figure 2
Figure 2
Development of mathematically optimized CombiWISE scale. (A) A bean plot (25) of variability in relative weights constructed from 200 permutations of training data using the genetic algorithm. (B) A bean plot of variability in weights for optimization across 500 permutations of training data using reduced set of clinical scales. (A,B) Black lines represent the individual weight results, red lines show the average of weights for each contributing scale, yellow areas represent non-parametric density curves of distribution of individual weights. (C) Mean relative weights and computing weights from the contributing clinical scales for CombiWISE calculation. Relative weights allow for comparing the level of contribution of individual scales while the computing weights are rescaled versions that produce a metric ranging from 0 to 100. (D) A bean plot of 500 permutations of validation data test-statistics for CombiWISE in addition to individual clinical scales and the Multiple Sclerosis Functional Composite (MSFC) scale. Black lines represent individual test statistics, red lines show average of test statistics for each scale, yellow areas represent non-parametric density curves of individual test statistics, blue lines correspond to cut-offs for 5% significance level tests.
Figure 3
Figure 3
Combinatorial weight-adjusted disability score (CombiWISE) correlates highly with other clinical scales and shows statistically significant progression in three independent cohorts. (A) Spearman correlations between CombiWISE and standard clinical scores (EDSS, SNRS, 25FW, 9HPT, SDMT, and MSFC) in the cross-sectional cohort of 303 untreated subjects with different types of MS, other inflammatory and non-inflammatory CNS conditions, and healthy volunteers. The Y-axis scales for 25FW and 9HTP are log2-transformed; ****p < 0.0001. (B) Longitudinal data for CombiWISE calculated from clinical scores collected every 6 months (Mo −12, Mo −6, and Mo 0) during the pre-treatment baseline for PPMS subjects in the IPPoMS trial and for a cohort of 29 untreated SPMS subjects showing statistically significant worsening of the clinical status over periods of 6 to 12 months in both PPMS and SPMS subjects. Statistical significance was determined by one-way ANOVA test on repeated measures. The red bars show mean for each group, *p < 0.05, **p < 0.01, ***p < 0.001, displayed that p-values were adjusted for multiple comparisons by Holm–Sidak test. (C) Longitudinal data for CombiWISE collected during the pre-treatment baseline of the IPPoMS trial show statistically significant increase in CombiWISE over 1 year. CombiWISE data collected on healthy subjects with a yearly follow-up visit show no overlap with the data of moderately to severely disabled PPMS patients, as well as no appreciable change over 1 year. Red bars represent the mean of the group. (D) Comparison of measured change and the overlap of values between IPPoMS and HV cohorts for the best-performing DTI biomarker (axial diffusivity of the medulla). Red bars represent the mean of the group.
Figure 4
Figure 4
Power analysis for the selected seven statistically significant variables. Power analysis shows number of required subjects per arm (accumulated sample size as effect size drops across bars) considering 80% nominal power, statistical significance level of 5% and either (A) parallel or (B) baseline vs. treatment group design for the 2-year study considering 30% (sum of black, gray, hatched bars), 40% (sum of gray and hatched bars), and 50% (hatched bars) drug effect.

References

    1. Wolinsky JS, Narayana PA, O’Connor P, Coyle PK, Ford C, Johnson K, et al. Glatiramer acetate in primary progressive multiple sclerosis: results of a multinational, multicenter, double-blind, placebo-controlled trial. Ann Neurol (2007) 61(1):14–24.10.1002/ana.21079
    1. Altmann DR, Jasperse B, Barkhof F, Beckmann K, Filippi M, Kappos LD, et al. Sample sizes for brain atrophy outcomes in trials for secondary progressive multiple sclerosis. Neurology (2009) 72(7):595–601.10.1212/01.wnl.0000335765.55346.fc
    1. Harrison DM, Caffo BS, Shiee N, Farrell JA, Bazin PL, Farrell SK, et al. Longitudinal changes in diffusion tensor-based quantitative MRI in multiple sclerosis. Neurology (2011) 76(2):179–86.10.1212/WNL.0b013e318206ca61
    1. Frost C, Kenward MG, Fox NC. Optimizing the design of clinical trials where the outcome is a rate. Can estimating a baseline rate in a run-in period increase efficiency? Stat Med (2008) 27(19):3717–31.10.1002/sim.3280
    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(1):479–89.10.1006/nimg.2002.1040
    1. Komori M, Lin YC, Cortese I, Blake A, Ohayon J, Cherup J, et al. Insufficient disease inhibition by intrathecal rituximab in progressive multiple sclerosis. Ann Clin Transl Neurol (2016) 3(3):166–79.10.1002/acn3.293
    1. SAS Institute Inc. SAS/STAT®9.22 User’s Guide. Cary, NC: SAS Institute; (2010).
    1. Kurtzke JF. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale (EDSS). Neurology (1983) 33(11):1444–52.10.1212/WNL.33.11.1444
    1. Sipe JC, Knobler RL, Braheny SL, Rice GP, Panitch HS, Oldstone MB. A neurologic rating scale (NRS) for use in multiple sclerosis. Neurology (1984) 34(10):1368–72.10.1212/WNL.34.10.1368
    1. Fischer JS, Rudick RA, Cutter GR, Reingold SC. The multiple sclerosis functional composite measure (MSFC): an integrated approach to MS clinical outcome assessment. National MS society clinical outcomes assessment task force. Mult Scler (1999) 5(4):244–50.10.1177/135245859900500409
    1. Hallett M. Transcranial magnetic stimulation: a primer. Neuron (2007) 55(2):187–99.10.1016/j.neuron.2007.06.026
    1. Duan Q, van Gelderen P, Duyn J. Improved Bloch-Siegert based B1 mapping by reducing off-resonance shift. NMR Biomed (2013) 26(9):1070–8.10.1002/nbm.2920
    1. Deoni SC. High-resolution T1 mapping of the brain at 3T with driven equilibrium single pulse observation of T1 with high-speed incorporation of RF field inhomogeneities (DESPOT1-HIFI). J Magn Reson Imaging (2007) 26(4):1106–11.10.1002/jmri.21130
    1. Shiee N, Bazin PL, Ozturk A, Reich DS, Calabresi PA, Pham DL. A topology-preserving approach to the segmentation of brain images with multiple sclerosis lesions. Neuroimage (2010) 49(2):1524–35.10.1016/j.neuroimage.2009.09.005
    1. Chang LC, Jones DK, Pierpaoli C. RESTORE: robust estimation of tensors by outlier rejection. Magn Reson Med (2005) 53(5):1088–95.10.1002/mrm.20426
    1. Landman BA, Farrell JA, Jones CK, Smith SA, Prince JL, Mori S. Effects of diffusion weighting schemes on the reproducibility of DTI-derived fractional anisotropy, mean diffusivity, and principal eigenvector measurements at 1.5T. Neuroimage (2007) 36(4):1123–38.10.1016/j.neuroimage.2007.02.056
    1. Scrucca L. GA: a package for genetic algorithms in R. J Stat Softw (2013) 53(4):1–37.10.18637/jss.v053.i04
    1. R Development Core Team. R: A Language and Environment for Statistical Computing. Vienna: R Foundation for Statistical Computing; (2016). Available from: ./
    1. Pinheiro J, Bates D. Mixed-Effects Models in S and S-PLUS. New York, NY: Springer-Verlag; (2000).
    1. Pinheiro J, Bates D, DebRoy S, Sarkar D, R Core Team nlme: Linear and Non-linear Mixed Effects Models. R package Version 3.1-117. (2014). Available from:
    1. Bock M, Brandt AU, Kuchenbecker J, Dorr J, Pfueller CF, Weinges-Evers N, et al. Impairment of contrast visual acuity as a functional correlate of retinal nerve fibre layer thinning and total macular volume reduction in multiple sclerosis. Br J Ophthalmol (2012) 96(1):62–7.10.1136/bjo.2010.193581
    1. Holland J. Adaptation in Natural and Artificial Systems. Ann Arbor: The University of Michigan Press; (1975).
    1. Goldberg D. Genetic Algorithms in Search, Optimization, and Machine Learning. Boston: Addison-Wesley; (1989).
    1. Sivanandam S, Deepa S. Introduction to Genetic Algorithms. Berlin: Springer-Verlag; (2008).
    1. Kampstra P. Beanplot: a boxplot alternative for visual comparison of distributions. J Stat Softw (2008) 28(1):1–9.10.18637/jss.v028.c01
    1. Ontaneda D, Fox RJ, Chataway J. Clinical trials in progressive multiple sclerosis: lessons learned and future perspectives. Lancet Neurol (2015) 14(2):208–23.10.1016/S1474-4422(14)70264-9
    1. Kapoor R, Furby J, Hayton T, Smith KJ, Altmann DR, Brenner R, et al. Lamotrigine for neuroprotection in secondary progressive multiple sclerosis: a randomised, double-blind, placebo-controlled, parallel-group trial. Lancet Neurol (2010) 9(7):681–8.10.1016/S1474-4422(10)70131-9
    1. Weinberger DR, Radulescu E. Finding the elusive psychiatric “lesion” with 21st-century neuroanatomy: a note of caution. Am J Psychiatry (2016) 173(1):27–33.10.1176/appi.ajp.2015.15060753
    1. Jones DK, Cercignani M. Twenty-five pitfalls in the analysis of diffusion MRI data. NMR Biomed (2010) 23(7):803–20.10.1002/nbm.1543
    1. Zhang J, Jones MV, McMahon MT, Mori S, Calabresi PA. In vivo and ex vivo diffusion tensor imaging of cuprizone-induced demyelination in the mouse corpus callosum. Magn Reson Med (2012) 67(3):750–9.10.1002/mrm.23032
    1. Jones DK, Knosche TR, Turner R. White matter integrity, fiber count, and other fallacies: the do’s and don’ts of diffusion MRI. Neuroimage (2013) 73:239–54.10.1016/j.neuroimage.2012.06.081
    1. Prentice RL. Surrogate endpoints in clinical trials: definition and operational criteria. Stat Med (1989) 8(4):431–40.10.1002/sim.4780080407
    1. Arnold DL. Evidence for neuroprotection and remyelination using imaging techniques. Neurology (2007) 68(22 Suppl 3):S83–90.10.1212/01.wnl.0000275237.28259.9d
    1. Khaleeli Z, Sastre-Garriga J, Ciccarelli O, Miller DH, Thompson AJ. Magnetisation transfer ratio in the normal appearing white matter predicts progression of disability over 1 year in early primary progressive multiple sclerosis. J Neurol Neurosurg Psychiatry (2007) 78(10):1076–82.10.1136/jnnp.2006.107565
    1. Khaleeli Z, Altmann DR, Cercignani M, Ciccarelli O, Miller DH, Thompson AJ. Magnetization transfer ratio in gray matter: a potential surrogate marker for progression in early primary progressive multiple sclerosis. Arch Neurol (2008) 65(11):1454–9.10.1001/archneur.65.11.1454
    1. Llufriu S, Kornak J, Ratiney H, Oh J, Brenneman D, Cree BA, et al. Magnetic resonance spectroscopy markers of disease progression in multiple sclerosis. JAMA Neurol (2014) 71(7):840–7.10.1001/jamaneurol.2014.895
    1. Goodkin DE, Rudick RA, VanderBrug Medendorp S, Greene T, Schwetz KM, Fischer J, et al. Low-dose (7.5 mg) oral methotrexate for chronic progressive multiple sclerosis. Design of a randomized, placebo-controlled trial with sample size benefits from a composite outcome variable including preliminary data on toxicity. Online J Curr Clin Trials (1992) 19:7723.
    1. Goodkin DE, Rudick RA, VanderBrug Medendorp S, Daughtry MM, Schwetz KM, Fischer J, et al. Low-dose (7.5 mg) oral methotrexate reduces the rate of progression in chronic progressive multiple sclerosis. Ann Neurol (1995) 37(1):30–40.10.1002/ana.410370108
    1. Rudick R, Antel J, Confavreux C, Cutter G, Ellison G, Fischer J, et al. Recommendations from the national multiple sclerosis society clinical outcomes assessment task force. Ann Neurol (1997) 42(3):379–82.10.1002/ana.410420318
    1. Kragt JJ, Thompson AJ, Montalban X, Tintore M, Rio J, Polman CH, et al. Responsiveness and predictive value of EDSS and MSFC in primary progressive MS. Neurology (2008) 70(13 Pt 2):1084–91.10.1212/01.wnl.0000288179.86056.e1

Source: PubMed

3
S'abonner