Pepinemab antibody blockade of SEMA4D in early Huntington's disease: a randomized, placebo-controlled, phase 2 trial

Andrew Feigin, Elizabeth E Evans, Terrence L Fisher, John E Leonard, Ernest S Smith, Alisha Reader, Vikas Mishra, Richard Manber, Kimberly A Walters, Lisa Kowarski, David Oakes, Eric Siemers, Karl D Kieburtz, Maurice Zauderer, Huntington Study Group SIGNAL investigators, Elise Kayson, Jody Goldstein, Richard Barbano, Karen Marder, Praveen Dayalu, Herminia Diana Rosas, Sandra Kostyk, John Kamholz, Brad Racette, Jee Bang, Daniel Claassen, Katherine McDonell, Stewart Factor, Francis Walker, Clarisse Goas, Joanne Wojcieszek, Lynn A Raymond, Jody Corey-Bloom, Victor Sung, Marissa Dean, Michael Geshwind, Alexandra Nelson, Samuel Frank, Kathrin LaFaver, Andrew Duker, Lawrence Elmer, Ali Samii, Yi-Han Lin, Sylvain Chouinard, Lauren Seeberger, Burton Scott, James Boyd, Nikolaus McFarland, Erin Furr Stimming, Oksana Suchowersky, Claudia Testa, Karen Anderson, Andrew Feigin, Elizabeth E Evans, Terrence L Fisher, John E Leonard, Ernest S Smith, Alisha Reader, Vikas Mishra, Richard Manber, Kimberly A Walters, Lisa Kowarski, David Oakes, Eric Siemers, Karl D Kieburtz, Maurice Zauderer, Huntington Study Group SIGNAL investigators, Elise Kayson, Jody Goldstein, Richard Barbano, Karen Marder, Praveen Dayalu, Herminia Diana Rosas, Sandra Kostyk, John Kamholz, Brad Racette, Jee Bang, Daniel Claassen, Katherine McDonell, Stewart Factor, Francis Walker, Clarisse Goas, Joanne Wojcieszek, Lynn A Raymond, Jody Corey-Bloom, Victor Sung, Marissa Dean, Michael Geshwind, Alexandra Nelson, Samuel Frank, Kathrin LaFaver, Andrew Duker, Lawrence Elmer, Ali Samii, Yi-Han Lin, Sylvain Chouinard, Lauren Seeberger, Burton Scott, James Boyd, Nikolaus McFarland, Erin Furr Stimming, Oksana Suchowersky, Claudia Testa, Karen Anderson

Abstract

SIGNAL is a multicenter, randomized, double-blind, placebo-controlled phase 2 study (no. NCT02481674) established to evaluate pepinemab, a semaphorin 4D (SEMA4D)-blocking antibody, for treatment of Huntington's disease (HD). The trial enrolled a total of 265 HD gene expansion carriers with either early manifest (EM, n = 179) or late prodromal (LP, n = 86) HD, randomized (1:1) to receive 18 monthly infusions of pepinemab (n = 91 EM, 41 LP) or placebo (n = 88 EM, 45 LP). Pepinemab was generally well tolerated, with a relatively low frequency of serious treatment-emergent adverse events of 5% with pepinemab compared to 9% with placebo, including both EM and LP participants. Coprimary efficacy outcome measures consisted of assessments within the EM cohort of (1) a two-item HD cognitive assessment family comprising one-touch stockings of Cambridge (OTS) and paced tapping (PTAP) and (2) clinical global impression of change (CGIC). The differences between pepinemab and placebo in mean change (95% confidence interval) from baseline at month 17 for OTS were -1.98 (-4.00, 0.05) (one-sided P = 0.028), and for PTAP 1.43 (-0.37, 3.23) (one-sided P = 0.06). Similarly, because a significant treatment effect was not observed for CGIC, the coprimary endpoint, the study did not meet its prespecified primary outcomes. Nevertheless, a number of other positive outcomes and post hoc subgroup analyses-including additional cognitive measures and volumetric magnetic resonance imaging and fluorodeoxyglucose-positron-emission tomography imaging assessments-provide rationale and direction for the design of a phase 3 study and encourage the continued development of pepinemab in patients diagnosed with EM HD.

Conflict of interest statement

Vaccinex employment and stock: E.E.E., T.L.F., J.E.L., E.S., V.M. and M.Z. Vaccinex patents and applications related to SEMA4D (USPTO nos. 8,816,058, 9,090,709, 10,800,853 and 7,919,594): E.E.E., T.L.F., E.S. and M.Z. IXICO employment and stock: R.M. Research grant support from HSG, Neurocrine, Uniqure and Vaccinex: P.D. Payments to Clintrex Research Corporation for provision of research services during the conduct of the trial: K.D.K. Research funding from Vaccinex, CHDI, HDSA, Roche/Genentech, UniQure, HSG/NBI and Cures within Reach, consulting role for Teva and on Speaker’s Bureau of Sunovion Pharmaceuticals: E.F.S. Consultation fees from Amylyz, Novartis, Sage, Teva and Uniqure, grant funding from uniQure, Roche/Genentech, Triplet Therapeutics and CHDI and salary support from HSG for serving as chair of DSMB of a study funded by Neurocrine and the virtual UHDRS study: S. Frank. The remaining authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Effects of pepinemab treatment on…
Fig. 1. Effects of pepinemab treatment on primary cognitive assessments in EM cohort B1.
a,b, Observed mean changes from baseline (BL) by treatment group over time for the mITT sample of EM cohort B1. a, OTS measures time to a correct response (averaged over all trials per visit). b, PTAP measures tapping consistency as the reciprocal of the average standard deviation of inter-tap interval durations following cessation of aural cues (over all trials per visit). a,b, Error bars show one standard error on either side of the mean, with sample sizes at each time point for each group listed above the profile lines.
Fig. 2. Pepinemab delays brain atrophy and…
Fig. 2. Pepinemab delays brain atrophy and restores loss of metabolic activity in EM subjects.
ad, Mean percentage changes from baseline by treatment group over time for the mITT sample of EM cohort B1 (PEPI, n = 90: PBO, n = 88) in vMRI measurement. a, Caudate BSI (atrophy); b, ventricular BSI (expansion); c, white matter (preservation); d, whole-brain BSI (atrophy). e, FDG–PET SUVR change from baseline to month 17 for each treatment group (mean and 1 s.d.) in each brain ROI for EM cohort B1. f. Treatment effect at month 17 calculated as difference between pepinemab (n = 40) and placebo groups (n = 36) as mean percentage change in SUVR. *P ≤ 0.05; exact two-sided P values for 15 brain regions (listed from top to bottom) are: extended frontal composite, 0.031; expanded cortical composite, 0.028; posterior cingulate, 0.008; lingual gyrus, 0.014; thalamus, 0.011; middle frontal gyrus, 0.033; occipital lobe, 0.029; precentral gyrus, 0.010, paracentral lobule, 0.014; post central gyrus, 0.028; precuneus cortex, 0.048; middle temporal gryus, 0.044; inferior temporal gyrus, 0.033; superior parietal, 0.050; superior temporal gyrus, 0.037; P values for all regions are shown in Extended Data Table 5. Analysis results were determined from MMRM of scheduled measurements at months 2, 6 and 17, with estimation of the difference in means between groups at month 17. P values are indicated (two-sided); as described in Methods, stated P values for all statistical tests, besides the coprimary efficacy analyses, were not corrected for multiplicity and are thus presented as nominal and not under alpha control. Error bars show one standard error on either side of the mean, with sample sizes at each time point for each group listed above the profile lines.
Fig. 3. Exploratory cognitive measures and post…
Fig. 3. Exploratory cognitive measures and post hoc subgroup analysis of baseline MoCA as a biomarker for treatment response in early HD.
ac, Observed mean changes from baseline by treatment group over time for the placebo groups alone of cohort B1 (red circles) and B2 (brown triangles) (a), for both placebo and pepinemab treatment groups of EM cohort B1 (b) and LP cohort B2 (c). Cognitive assessments stratified by baseline MoCA scores of <26 and 26–30 for assessments of HD–CAB index (d), PTAP (e) and OTS (f). Error bars in each panel show one standard error on either side of the mean, with sample sizes at each time point for each group listed above the profile lines. PEPI (blue squares) and PBO (red circles) in EM cohort B1, and PEPI (teal diamonds) and PBO (brown triangles) in LP cohort B2.
Fig. 4. Effects of pepinemab treatment on…
Fig. 4. Effects of pepinemab treatment on CGIC in EM cohort B1.
a, Observed categorical CGIC values at visit 17 for the entire EM cohort B1 CGIC analysis population, and in two subgroups stratified by baseline TFC value (that is, 11 and 12–13). b, Observed categorical CGIC in the subgroups stratified by TFC value over the study duration. The CGIC is a seven-point Likert scale, ranging from very much worse (−3) to very much improved (+3). Values were set to −3 following a patient death adjudged by a blinded data review committee to be related to Huntington’s disease. P values determined by one-sided Fisher’s exact test; odds ratio (95% CI) are also shown.
Extended Data Fig. 1
Extended Data Fig. 1
CONSORT diagram indicating participant numbers and disposition.
Extended Data Fig. 2. Pepinemab is detected…
Extended Data Fig. 2. Pepinemab is detected in CSF at expected level for target engagement.
a. Drug concentration in CSF of subjects treated with pepinemab. Mean+SEM is shown; dotted lines indicate target concentration (~100-300 ng/ml). b. Concentration of soluble SEMA4D (sSEMA4D) in CSF, mean+SEM are shown for each treatment group (n = 26 PEPI, n = 28 PBO). *** indicates statistical significance, p = 0.000000001855. As seen in previous trials,, levels of total soluble SEMA4D (including complex of drug bound to target) increased 1.7-fold upon dosing due to the increased half-life of the pepinemab/SEMA4D complex in subjects treated with pepinemab compared to those treated with placebo (average of 5.9 vs 3.5 ng/ml respectively, p < 0.001), demonstrating evidence of target engagement in CSF. The mean (SD) observed maximum serum concentration (Cmax) after all infusions post visit 12 in Cohort B overall was 218 (115) µg/mL and the AUCtau over the dosing interval was 68,900 (14,447) µg*hr/mL. Based on the empirical Bayesian estimates of the pharmacokinetic parameters, the terminal elimination half-lives for Cohorts B1 and B2 were calculated to be approximately 25 and 23 days, respectively. In general, the clearance of pepinemab was low and the volume of distribution small, which is common to other therapeutic monoclonal antibodies and similar to observations in previously completed studies with pepinemab,.

References

    1. Glidden, A. et al. in Movement Disorders Vol. 32 (ed A. Jon Stoessl), Supplement 2, S183 (Wiley, 2017).
    1. Simpson JA, Lovecky D, Kogan J, Vetter LA, Yohrling GJ. Survey of the Huntington’s disease patient and caregiver community reveals most impactful symptoms and treatment needs. J. Huntingt. Dis. 2016;5:395–403. doi: 10.3233/JHD-160228.
    1. Rub U, et al. Huntington’s disease (HD): the neuropathology of a multisystem neurodegenerative disorder of the human brain. Brain Pathol. 2016;26:726–740. doi: 10.1111/bpa.12426.
    1. Wilton DK, Stevens B. The contribution of glial cells to Huntington’s disease pathogenesis. Neurobiol. Dis. 2020;143:104963. doi: 10.1016/j.nbd.2020.104963.
    1. Basile JR, Gavard J, Gutkind JS. Plexin-B1 utilizes RHOA and ROK to promote the integrin-dependent activation of AKT and ERK, and endothelial cell motility. J. Biol. Chem. 2007;282:34888–34895. doi: 10.1074/jbc.M705467200.
    1. Liang X, Draghi NA, Resh MD. Signaling from integrins to Fyn to Rho family GTPases regulates morphologic differentiation of oligodendrocytes. J. Neurosci. 2004;24:7140–7149. doi: 10.1523/JNEUROSCI.5319-03.2004.
    1. Tamagnone L, et al. Plexins are a large family of receptors for transmembrane, secreted, and GPI-anchored semaphorins in vertebrates. Cell. 1999;99:71–80. doi: 10.1016/S0092-8674(00)80063-X.
    1. Denis HL, Lauruol F, Cicchetti F. Are immunotherapies for Huntington’s disease a realistic option? Mol. Psychiatry. 2019;24:364–377. doi: 10.1038/s41380-018-0021-9.
    1. Toguchi, M., Gonzalez, D., Furukawa, S. & Inagaki, S. Involvement of Sema4D in the control of microglia activation. Neurochem. Int. 55, 573–580 (2009).
    1. Southwell AL, et al. Anti-semaphorin 4D immunotherapy ameliorates neuropathology and some cognitive impairment in the YAC128 mouse model of Huntington disease. Neurobiol. Dis. 2015;76:46–56. doi: 10.1016/j.nbd.2015.01.002.
    1. Chapoval, S. P., Vadasz, Z., Chapoval, A. I. & Toubi, E. Semaphorins 4A and 4D in chronic inflammatory diseases. Inflamm. Res.66, 111–117 (2016).
    1. Wu M, Li J, Gao Q, Ye F. The role for Sema4D/CD100 as a therapeutic target for tumor microenvironments and for autoimmune, neuroimmune and bone diseases. Expert Opin. Ther. Targets. 2016;20:885–901. doi: 10.1517/14728222.2016.1139083.
    1. Smith ES, et al. SEMA4D compromises blood-brain barrier, activates microglia, and inhibits remyelination in neurodegenerative disease. Neurobiol. Dis. 2014;73:254–268. doi: 10.1016/j.nbd.2014.10.008.
    1. Okuno T, et al. Roles of SEMA4D-plexin-B1 interactions in the central nervous system for pathogenesis of experimental autoimmune encephalomyelitis. J. Immunol. 2010;184:1499–1506. doi: 10.4049/jimmunol.0903302.
    1. Giraudon P, et al. Semaphorin CD100 from activated T lymphocytes induces process extension collapse in oligodendrocytes and death of immature neural cells. J. Immunol. 2004;172:1246–1255. doi: 10.4049/jimmunol.172.2.1246.
    1. Giraudon P, Vincent P, Vuaillat C. T-cells in neuronal injury and repair: semaphorins and related T-cell signals. Neuromolecular. Med. 2005;7:207–216. doi: 10.1385/NMM:7:3:207.
    1. Chen WW, Zhang X, Huang WJ. Role of neuroinflammation in neurodegenerative diseases (Review) Mol. Med. Rep. 2016;13:3391–3396. doi: 10.3892/mmr.2016.4948.
    1. Patnaik A, et al. Safety, pharmacokinetics, and pharmacodynamics of a humanized anti-semaphorin 4D antibody, in a first-in-human study of patients with advanced solid tumors. Clin. Cancer Res. 2015;22:827–836. doi: 10.1158/1078-0432.CCR-15-0431.
    1. LaGanke C, et al. Safety/tolerability of the anti-semaphorin 4D antibody VX15/2503 in a randomized phase 1 trial. Neurol. Neuroimmunol. Neuroinflamm. 2017;4:e367. doi: 10.1212/NXI.0000000000000367.
    1. Shafique, M. et al. A Phase 1b/2 study of pepinemab in combination with avelumab in advanced non-small cell lung cancer. Clin. Cancer Res.27, 3630–3640 (2021).
    1. Tabrizi SJ, et al. Potential endpoints for clinical trials in premanifest and early Huntington’s disease in the TRACK-HD study: analysis of 24 month observational data. Lancet Neurol. 2012;11:42–53. doi: 10.1016/S1474-4422(11)70263-0.
    1. Tabrizi SJ, et al. Predictors of phenotypic progression and disease onset in premanifest and early-stage Huntington’s disease in the TRACK-HD study: analysis of 36-month observational data. Lancet Neurol. 2013;12:637–649. doi: 10.1016/S1474-4422(13)70088-7.
    1. Reilmann R, et al. Safety and efficacy of pridopidine in patients with Huntington’s disease (PRIDE-HD): a phase 2, randomised, placebo-controlled, multicentre, dose-ranging study. Lancet Neurol. 2019;18:165–176. doi: 10.1016/S1474-4422(18)30391-0.
    1. Tabrizi SJ, et al. Biological and clinical manifestations of Huntington’s disease in the longitudinal TRACK-HD study: cross-sectional analysis of baseline data. Lancet Neurol. 2009;8:791–801. doi: 10.1016/S1474-4422(09)70170-X.
    1. Wilson H, De Micco R, Niccolini F, Politis M. Molecular imaging markers to track Huntington’s disease pathology. Front. Neurol. 2017;8:11. doi: 10.3389/fneur.2017.00011.
    1. Tang CC, et al. Metabolic network as a progression biomarker of premanifest Huntington’s disease. J. Clin. Invest. 2013;123:4076–4088. doi: 10.1172/JCI69411.
    1. Feigin A, et al. Thalamic metabolism and symptom onset in preclinical Huntington’s disease. Brain. 2007;130:2858–2867. doi: 10.1093/brain/awm217.
    1. Landau SM, et al. Associations between cognitive, functional, and FDG-PET measures of decline in AD and MCI. Neurobiol. Aging. 2011;32:1207–1218. doi: 10.1016/j.neurobiolaging.2009.07.002.
    1. Hanseeuw BJ, et al. Fluorodeoxyglucose metabolism associated with tau-amyloid interaction predicts memory decline. Ann. Neurol. 2017;81:583–596. doi: 10.1002/ana.24910.
    1. Stout JC, et al. HD-CAB: a cognitive assessment battery for clinical trials in Huntington’s disease 1,2,3. Mov. Disord. 2014;29:1281–1288. doi: 10.1002/mds.25964.
    1. Watkins LH, et al. Impaired planning but intact decision making in early Huntington’s disease: implications for specific fronto-striatal pathology. Neuropsychologia. 2000;38:1112–1125. doi: 10.1016/S0028-3932(00)00028-2.
    1. Rowe KC, et al. Self-paced timing detects and tracks change in prodromal Huntington disease. Neuropsychology. 2010;24:435–442. doi: 10.1037/a0018905.
    1. Smith, A. Symbol Digit Modalities Test (Western Psychological Services, 1973).
    1. Ekman P, Friesen WV. Measuring facial movement. Environ. Psychol. Nonverbal Behav. 1976;1:56–75. doi: 10.1007/BF01115465.
    1. Johnson SA, et al. Beyond disgust: impaired recognition of negative emotions prior to diagnosis in Huntington’s disease. Brain. 2007;130:1732–1744. doi: 10.1093/brain/awm107.
    1. Brandt, J. & Benedict, R. H. Hopkins Verbal Learning Test–Revised: Professional Manual (Psychological Assessment Resources, 2001).
    1. Reitan RM. Validity of the Trail Making Test as an indicator of organic brain damage. Percept. Mot. Skills. 1958;8:271–276. doi: 10.2466/pms.1958.8.3.271.
    1. Hochberg Y. A sharper Bonferroni procedure for multiple tests of significance. Biometrika. 1988;75:800–802. doi: 10.1093/biomet/75.4.800.
    1. Cognition, C. CANTABeclipse. Test Administration Guide/Manual version 3.0.0 (Cambridge Cognition Ltd, 2006).
    1. Guy, W. Assessment Manual for Psychopharmacology (US Government Printing Office, 1976).
    1. Reilmann R, Schubert R. Motor outcome measures in Huntington disease clinical trials. Handb. Clin. Neurol. 2017;144:209–225. doi: 10.1016/B978-0-12-801893-4.00018-3.
    1. Freeborough PA, Fox NC. The boundary shift integral: an accurate and robust measure of cerebral volume changes from registered repeat MRI. IEEE Trans. Med. Imaging. 1997;16:623–629. doi: 10.1109/42.640753.
    1. Schobel, S. Preliminary results from GENERATION HD1, a phase III trial of tominersen in individuals with manifest HD. In CHDI 16th Annual HD Therapeutics Conference (2021).
    1. Jutten RJ, et al. Monthly at-home computerized cognitive testing to detect diminished practice effects in preclinical Alzheimer’s disease. Front. Aging Neurosci. 2021;13:800126. doi: 10.3389/fnagi.2021.800126.
    1. Samaroo A, et al. Diminished Learning Over Repeated Exposures (LORE) in preclinical Alzheimer’s disease. Alzheimers Dement. (Amst.) 2020;12:e12132.
    1. Baudic S, et al. Cognitive impairment related to apathy in early Huntington’s disease. Dement. Geriatr. Cogn. Disord. 2006;21:316–321. doi: 10.1159/000091523.
    1. Kremer H, Group HS. Unified Huntington’s disease rating scale: reliability and consistency. Mov. Disord. 1996;11:136–142. doi: 10.1002/mds.870110204.
    1. Stout JC, et al. Evaluation of longitudinal 12 and 24 month cognitive outcomes in premanifest and early Huntington’s disease. J. Neurol. Neurosurg. Psychiatry. 2012;83:687–694. doi: 10.1136/jnnp-2011-301940.
    1. Tabrizi SJ, et al. Biological and clinical changes in premanifest and early stage Huntington’s disease in the TRACK-HD study: the 12-month longitudinal analysis. Lancet Neurol. 2011;10:31–42. doi: 10.1016/S1474-4422(10)70276-3.
    1. Posner K, et al. The Columbia-Suicide Severity Rating Scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults. Am. J. Psychiatry. 2011;168:1266–1277. doi: 10.1176/appi.ajp.2011.10111704.
    1. Khosravi M, et al. 18F-FDG is a superior indicator of cognitive performance compared to 18F-florbetapir in Alzheimer’s disease and mild cognitive impairment evaluation: a global quantitative analysis. J. Alzheimers Dis. 2019;70:1197–1207. doi: 10.3233/JAD-190220.
    1. Johnson EB, et al. Dynamics of cortical degeneration over a decade in Huntington’s disease. Biol. Psychiatry. 2021;89:807–816. doi: 10.1016/j.biopsych.2020.11.009.
    1. Zimmer ER, et al. [(18)F]FDG PET signal is driven by astroglial glutamate transport. Nat. Neurosci. 2017;20:393–395. doi: 10.1038/nn.4492.
    1. Boussicault L, et al. Impaired brain energy metabolism in the BACHD mouse model of Huntington’s disease: critical role of astrocyte-neuron interactions. J. Cereb. Blood Flow. Metab. 2014;34:1500–1510. doi: 10.1038/jcbfm.2014.110.
    1. Polyzos AA, et al. Metabolic reprogramming in astrocytes distinguishes region-specific neuronal susceptibility in Huntington mice. Cell Metab. 2019;29:1258–1273. doi: 10.1016/j.cmet.2019.03.004.
    1. Clark, I. C. et al. Barcoded viral tracing of single-cell interactions in central nervous system inflammation. Science372, eabf1230(2021).
    1. Zhang Y, et al. Indexing disease progression at study entry with individuals at-risk for Huntington disease. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2011;156B:751–763. doi: 10.1002/ajmg.b.31232.
    1. Fisher TL, et al. Generation and preclinical characterization of an antibody specific for SEMA4D. mAbs. 2016;8:150–162. doi: 10.1080/19420862.2015.1102813.
    1. Labrijn AF, et al. Therapeutic IgG4 antibodies engage in Fab-arm exchange with endogenous human IgG4 in vivo. Nat. Biotechnol. 2009;27:767–771. doi: 10.1038/nbt.1553.
    1. Leonard JE, et al. Nonclinical safety evaluation of VX15/2503, a humanized IgG4 anti-SEMA4D antibody. Mol. Cancer Ther. 2015;14:964–972. doi: 10.1158/1535-7163.MCT-14-0924.
    1. Feigin A, et al. Functional decline in Huntington’s disease. Mov. Disord. 1995;10:211–214. doi: 10.1002/mds.870100213.
    1. Siesling S, van Vugt JP, Zwinderman KA, Kieburtz K, Roos RA. Unified Huntington’s disease rating scale: a follow up. Mov. Disord. 1998;13:915–919. doi: 10.1002/mds.870130609.
    1. Callaghan J, et al. Reliability and factor structure of the Short Problem Behaviors Assessment for Huntington’s disease (PBA-s) in the TRACK-HD and REGISTRY studies. J. Neuropsychiatry Clin. Neurosci. 2015;27:59–64. doi: 10.1176/appi.neuropsych.13070169.
    1. Wolz R, et al. LEAP: learning embeddings for atlas propagation. Neuroimage. 2010;49:1316–1325. doi: 10.1016/j.neuroimage.2009.09.069.
    1. Wolz R, et al. Measurement of hippocampal atrophy using 4D graph-cut segmentation: application to ADNI. Neuroimage. 2010;52:109–118. doi: 10.1016/j.neuroimage.2010.04.006.
    1. Ledig C, et al. Robust whole-brain segmentation: application to traumatic brain injury. Med. Image Anal. 2015;21:40–58. doi: 10.1016/j.media.2014.12.003.
    1. McGarry A, et al. A randomized, double-blind, placebo-controlled trial of coenzyme Q10 in Huntington disease. Neurology. 2017;88:152–159. doi: 10.1212/WNL.0000000000003478.

Source: PubMed

3
Abonner