51
|
Lee JM, Huang Y, Orth M, Gillis T, Siciliano J, Hong E, Mysore JS, Lucente D, Wheeler VC, Seong IS, McLean ZL, Mills JA, McAllister B, Lobanov SV, Massey TH, Ciosi M, Landwehrmeyer GB, Paulsen JS, Dorsey ER, Shoulson I, Sampaio C, Monckton DG, Kwak S, Holmans P, Jones L, MacDonald ME, Long JD, Gusella JF. Genetic modifiers of Huntington disease differentially influence motor and cognitive domains. Am J Hum Genet 2022; 109:885-899. [PMID: 35325614 DOI: 10.1016/j.ajhg.2022.03.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Genome-wide association studies (GWASs) of Huntington disease (HD) have identified six DNA maintenance gene loci (among others) as modifiers and implicated a two step-mechanism of pathogenesis: somatic instability of the causative HTT CAG repeat with subsequent triggering of neuronal damage. The largest studies have been limited to HD individuals with a rater-estimated age at motor onset. To capitalize on the wealth of phenotypic data in several large HD natural history studies, we have performed algorithmic prediction by using common motor and cognitive measures to predict age at other disease landmarks as additional phenotypes for GWASs. Combined with imputation with the Trans-Omics for Precision Medicine reference panel, predictions using integrated measures provided objective landmark phenotypes with greater power to detect most modifier loci. Importantly, substantial differences in the relative modifier signal across loci, highlighted by comparing common modifiers at MSH3 and FAN1, revealed that individual modifier effects can act preferentially in the motor or cognitive domains. Individual components of the DNA maintenance modifier mechanisms may therefore act differentially on the neuronal circuits underlying the corresponding clinical measures. In addition, we identified additional modifier effects at the PMS1 and PMS2 loci and implicated a potential second locus on chromosome 7. These findings indicate that broadened discovery and characterization of HD genetic modifiers based on additional quantitative or qualitative phenotypes offers not only the promise of in-human validated therapeutic targets but also a route to dissecting the mechanisms and cell types involved in both the somatic instability and toxicity components of HD pathogenesis.
Collapse
|
52
|
Hawellek DJ, Garces P, Meghdadi AH, Waninger S, Smith A, Manchester M, Schobel SA, Hipp JF. Changes in brain activity with tominersen in early-manifest Huntington’s disease. Brain Commun 2022; 4:fcac149. [PMID: 35774187 PMCID: PMC9237739 DOI: 10.1093/braincomms/fcac149] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/08/2022] [Accepted: 06/07/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
It is unknown whether alterations in EEG brain activity caused by Huntington’s disease may be responsive to huntingtin-lowering treatment. We analysed EEG recordings of 46 patients (mean age = 47.02 years; standard deviation = 10.19 years; 18 female) with early-manifest Stage 1 Huntington’s disease receiving the huntingtin-lowering antisense oligonucleotide tominersen for 4 months or receiving placebo as well as 39 healthy volunteers (mean age = 44.48 years; standard deviation = 12.94; 22 female) not receiving treatment. Patients on tominersen showed increased resting-state activity within a 4–8 Hz frequency range compared with patients receiving placebo (cluster-based permutation test, P < 0.05). The responsive frequency range overlapped with EEG activity that was strongly reduced in Huntington’s disease compared with healthy controls (cluster-based permutation test, P < 0.05). The underlying mechanisms of the observed treatment-related increase are unknown and may reflect neural plasticity as a consequence of the molecular pathways impacted by tominersen treatment.
Hawellek et al. report that patients with Huntington’s disease treated with the huntingtin-lowering antisense oligonucleotide tominersen exhibited increased EEG power in the theta/alpha frequency range. The underlying mechanisms of the observed changes are unknown and may reflect neural plasticity as a consequence of the molecular pathways impacted by tominersen treatment.
Collapse
Affiliation(s)
- D J Hawellek
- Roche, Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. , Basel 4070 , Switzerland
| | - P Garces
- Roche, Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. , Basel 4070 , Switzerland
| | - A H Meghdadi
- Advanced Brain Monitoring Inc. , Carlsbad, CA 92008 , USA
| | - S Waninger
- Advanced Brain Monitoring Inc. , Carlsbad, CA 92008 , USA
| | - A Smith
- Ionis Pharmaceuticals Inc. , Carlsbad, CA 92010 , USA
| | - M Manchester
- Roche, Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. , Basel 4070 , Switzerland
| | - S A Schobel
- F. Hoffmann-La Roche Ltd , Basel 4070 , Switzerland
| | - J F Hipp
- Roche, Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. , Basel 4070 , Switzerland
| |
Collapse
|
53
|
Lemercier P, Cleret de Langavant L, Hamet Bagnou J, Youssov K, Lemoine L, Audureau E, Massart R, Bachoud-Lévi AC. Self-Reported Social Relationship Capacities Predict Motor, Functional and Cognitive Decline in Huntington’s Disease. J Pers Med 2022; 12:jpm12020174. [PMID: 35207662 PMCID: PMC8879028 DOI: 10.3390/jpm12020174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/10/2022] Open
Abstract
Huntington’s Disease (HD) is an inherited neurodegenerative disease characterized by a combination of motor, cognitive, and behavioral disorders. The social and behavioral symptoms observed in HD patients impact their quality of life and probably explain their relational difficulties, conflicts, and social withdrawal. In this study, we described the development of the Social Relationship Self-Questionnaire (SRSQ), a self-reporting questionnaire that assesses how HD patients perceived their social relationships. The scale was proposed for 66 HD patients at an early stage of the disease, 32 PreHD patients (individuals carrying the mutant gene without motor symptoms), and 66 controls. The HD patients were included in a prospective longitudinal follow-up for an average of 1.07 years with motor, functional, cognitive, and behavioral assessments. Based on the HD patients’ answers at baseline, we identified two domains in the SRSQ. The first domain was related to social motivation and correlated with cognitive performance. The second domain was related to emotional insight and correlated with behavioral symptoms such as apathy, anxiety, and irritability. We discovered that both SRSQ domain scores at baseline predicted future motor, functional, and cognitive decline in HD.
Collapse
Affiliation(s)
- Pablo Lemercier
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL University, 75005 Paris, France; (P.L.); (L.C.d.L.); (J.H.B.); (K.Y.); (L.L.); (R.M.)
- Equipe NeuroPsychologie Interventionnelle, Institut Mondor de Recherche Biomédicale, INSERM U955, University Paris Est Créteil, 94000 Créteil, France
- Centre National de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, AP-HP, 94000 Créteil, France
- NeurATRIS, 94000 Créteil, France
| | - Laurent Cleret de Langavant
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL University, 75005 Paris, France; (P.L.); (L.C.d.L.); (J.H.B.); (K.Y.); (L.L.); (R.M.)
- Equipe NeuroPsychologie Interventionnelle, Institut Mondor de Recherche Biomédicale, INSERM U955, University Paris Est Créteil, 94000 Créteil, France
- Centre National de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, AP-HP, 94000 Créteil, France
- NeurATRIS, 94000 Créteil, France
| | - Jennifer Hamet Bagnou
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL University, 75005 Paris, France; (P.L.); (L.C.d.L.); (J.H.B.); (K.Y.); (L.L.); (R.M.)
- Equipe NeuroPsychologie Interventionnelle, Institut Mondor de Recherche Biomédicale, INSERM U955, University Paris Est Créteil, 94000 Créteil, France
- Centre National de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, AP-HP, 94000 Créteil, France
- NeurATRIS, 94000 Créteil, France
| | - Katia Youssov
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL University, 75005 Paris, France; (P.L.); (L.C.d.L.); (J.H.B.); (K.Y.); (L.L.); (R.M.)
- Centre National de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, AP-HP, 94000 Créteil, France
- NeurATRIS, 94000 Créteil, France
| | - Laurie Lemoine
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL University, 75005 Paris, France; (P.L.); (L.C.d.L.); (J.H.B.); (K.Y.); (L.L.); (R.M.)
- Equipe NeuroPsychologie Interventionnelle, Institut Mondor de Recherche Biomédicale, INSERM U955, University Paris Est Créteil, 94000 Créteil, France
- Centre National de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, AP-HP, 94000 Créteil, France
- NeurATRIS, 94000 Créteil, France
| | - Etienne Audureau
- Clinical Epidemiology and Ageing, Service de Santé Publique, Henri Mondor Hospital, AP-HP, 94000 Créteil, France;
| | - Renaud Massart
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL University, 75005 Paris, France; (P.L.); (L.C.d.L.); (J.H.B.); (K.Y.); (L.L.); (R.M.)
- Equipe NeuroPsychologie Interventionnelle, Institut Mondor de Recherche Biomédicale, INSERM U955, University Paris Est Créteil, 94000 Créteil, France
- Centre National de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, AP-HP, 94000 Créteil, France
- NeurATRIS, 94000 Créteil, France
| | - Anne-Catherine Bachoud-Lévi
- Département d’Etudes Cognitives, Ecole Normale Supérieure, PSL University, 75005 Paris, France; (P.L.); (L.C.d.L.); (J.H.B.); (K.Y.); (L.L.); (R.M.)
- Equipe NeuroPsychologie Interventionnelle, Institut Mondor de Recherche Biomédicale, INSERM U955, University Paris Est Créteil, 94000 Créteil, France
- Centre National de Référence Maladie de Huntington, Service de Neurologie, Hôpital Henri Mondor-Albert Chenevier, AP-HP, 94000 Créteil, France
- NeurATRIS, 94000 Créteil, France
- Correspondence:
| |
Collapse
|
54
|
Migliore S, D’Aurizio G, Scaricamazza E, Maffi S, Ceccarelli C, Ristori G, Romano S, Castaldo A, Fichera M, Curcio G, Squitieri F. Cognitive Reserve in Early Manifest Huntington Disease Patients: Leisure Time Is Associated with Lower Cognitive and Functional Impairment. J Pers Med 2022; 12:jpm12010036. [PMID: 35055351 PMCID: PMC8777615 DOI: 10.3390/jpm12010036] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/19/2021] [Accepted: 12/30/2021] [Indexed: 12/15/2022] Open
Abstract
We focused on Cognitive Reserve (CR) in patients with early Huntington Disease (HD) and investigated whether clinical outcomes might be influenced by lifetime intellectual enrichment over time. CR was evaluated by means of the Cognitive Reserve Index questionnaire (CRIq), an internationally validated scale which includes three sections: education, working activity, and leisure time. The clinical HD variables were quantified at three different time points (baseline-t0, 1 year follow up-t1 and 2 years follow up-t2) as per the Unified Huntington’s Disease Rating Scale (UHDRS), an internationally standardized and validated scale including motor, cognitive, functional and behavioral assays. Our sample consisted of 75 early manifest patients, withclinical stage scored according to the Total Functional Capacity (TFC) scale. Our correlational analysis highlighted a significant inverse association between CRIq leisure time (CRIq_LA) and longitudinal functional impairment (namely, the differential TFC score between t2 and t0 or ΔTFC) (p < 0.05), and the multidimensional progression of HD as measured by the composite UHDRS (cUHDRS, p < 0.01). CRIq_LA was significantly and positively associated with better cognitive performances at all time points (p < 0.05). Our results suggest that higher is the CRIq_LA, milder is the progression of HD in terms of functional, multidimensional and cognitive outcome.
Collapse
Affiliation(s)
- Simone Migliore
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza Hospital, 71013 San Giovanni Rotondo, Italy; (S.M.); (E.S.); (S.M.)
| | - Giulia D’Aurizio
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.D.); (G.C.)
| | - Eugenia Scaricamazza
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza Hospital, 71013 San Giovanni Rotondo, Italy; (S.M.); (E.S.); (S.M.)
| | - Sabrina Maffi
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza Hospital, 71013 San Giovanni Rotondo, Italy; (S.M.); (E.S.); (S.M.)
| | - Consuelo Ceccarelli
- Italian League for Research on Huntington and Related Diseases Foundation, 00185 Rome, Italy;
| | - Giovanni Ristori
- Centre for Experimental Neurological Therapies, Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, S. Andrea Hospital, Sapienza University, 00189 Rome, Italy; (G.R.); (S.R.)
| | - Silvia Romano
- Centre for Experimental Neurological Therapies, Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, S. Andrea Hospital, Sapienza University, 00189 Rome, Italy; (G.R.); (S.R.)
| | - Anna Castaldo
- Department of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (A.C.); (M.F.)
| | - Mario Fichera
- Department of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (A.C.); (M.F.)
| | - Giuseppe Curcio
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.D.); (G.C.)
| | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo Della Sofferenza Hospital, 71013 San Giovanni Rotondo, Italy; (S.M.); (E.S.); (S.M.)
- Correspondence: or
| |
Collapse
|
55
|
Drew CJG, Busse M. Considerations for clinical trial design and conduct in the evaluation of novel advanced therapeutics in neurodegenerative disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 166:235-279. [PMID: 36424094 DOI: 10.1016/bs.irn.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The recent advances in the development of potentially disease modifying cell and gene therapies for neurodegenerative disease has resulted in the production of a number of promising novel therapies which are now moving forward to clinical evaluation. The robust evaluation of these therapies pose a significant number of challenges when compared to more traditional evaluations of pharmacotherapy, which is the current mainstay of neurodegenerative disease symptom management. Indeed, there is an inherent complexity in the design and conduct of these trials at multiple levels. Here we discuss specific aspects requiring consideration in the context of investigating novel cell and gene therapies for neurodegenerative disease. This extends to overarching trial designs that could be employed and the factors that underpin design choices such outcome assessments, participant selection and methods for delivery of cell and gene therapies. We explore methods of data collection that may improve efficiency in trials of cell and gene therapy to maximize data sharing and collaboration. Lastly, we explore some of the additional context beyond efficacy evaluations that should be considered to ensure implementation across relevant healthcare settings.
Collapse
Affiliation(s)
- Cheney J G Drew
- Centre For Trials Research, Cardiff University, Cardiff, United Kingdom; Brain Repair and Intracranial Neurotherapeutics Unit (BRAIN), College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom.
| | - Monica Busse
- Centre For Trials Research, Cardiff University, Cardiff, United Kingdom; Brain Repair and Intracranial Neurotherapeutics Unit (BRAIN), College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
56
|
McLauchlan DJ, Lancaster T, Craufurd D, Linden DEJ, Rosser AE. Different depression: motivational anhedonia governs antidepressant efficacy in Huntington's disease. Brain Commun 2022; 4:fcac278. [PMID: 36440100 PMCID: PMC9683390 DOI: 10.1093/braincomms/fcac278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/13/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Depression is more common in neurodegenerative diseases such as Huntington's disease than the general population. Antidepressant efficacy is well-established for depression within the general population: a recent meta-analysis showed serotonin norepinephrine reuptake inhibitors, tricyclic antidepressants and mirtazapine outperformed other antidepressants. Despite the severe morbidity, antidepressant choice in Huntington's disease is based on Class IV evidence. We used complementary approaches to determine treatment choice for depression in Huntington's disease: propensity score analyses of antidepressant treatment outcome using the ENROLL-HD data set, and a dissection of the cognitive mechanisms underlying depression in Huntington's disease using a cognitive battery based on the Research Domain Criteria for Depression. Study 1 included ENROLL-HD 5486 gene-positive adult patients started on an antidepressant medication for depression. Our outcome measures were depression (Hospital Anxiety and Depression Scale or Problem Behaviours Assessment 'Depressed Mood' item) at first follow-up (primary outcome) and all follow-ups (secondary outcome). The intervention was antidepressant class. We used Svyglm&Twang in R to perform propensity scoring, using known variables (disease progression, medical comorbidity, psychiatric morbidity, sedatives, number of antidepressants, demographics and antidepressant contraindications) to determine the probability of receiving different antidepressants (propensity score) and then included the propensity score in a model of treatment efficacy. Study 2 recruited 51 gene-positive adult patients and 26 controls from the South Wales Huntington's Disease Management Service. Participants completed a motor assessment, in addition to measures of depression and apathy, followed by tasks measuring consummatory anhedonia, motivational anhedonia, learning from reward and punishment and reaction to negative outcome. We used generalised linear models to determine the association between task performance and depression scores. Study 1 showed selective serotonin reuptake inhibitors outperformed serotonin norepinephrine reuptake inhibitors on the primary outcome (P = 0.048), whilst both selective serotonin reuptake inhibitors (P = 0.00069) and bupropion (P = 0.0045) were superior to serotonin norepinephrine reuptake inhibitors on the secondary outcome. Study 2 demonstrated an association between depression score and effort for reward that was not explained by apathy. No other mechanisms were associated with depression score. We found that selective serotonin reuptake inhibitors and bupropion outperform serotonin norepinephrine reuptake inhibitors at alleviating depression in Huntington's disease. Moreover, motivational anhedonia appears the most significant mechanism underlying depression in Huntington's disease. Bupropion is improves motivational anhedonia and has a synergistic effect with selective serotonin reuptake inhibitors. This work provides the first large-scale, objective evidence to determine treatment choice for depression in Huntington's disease, and provides a model for determining antidepressant efficacy in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Duncan James McLauchlan
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK.,Department of Neurology, Morriston Hospital, Swansea Bay University Health Board, Swansea SA6 6NL, UK
| | - Thomas Lancaster
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK.,Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff CF24 4HQ, UK.,Department of Psychology, University of Bath, Bath BA2 7AY, UK
| | - David Craufurd
- Manchester Center for Genomic Medicine, Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Center, Manchester M13 9PL, UK.,St. Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Center, Manchester M13 9WL, UK
| | - David E J Linden
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK.,Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff CF24 4HQ, UK.,Department of Psychology, University of Bath, Bath BA2 7AY, UK.,School for Mental Health and Neuroscience, Fac. Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Anne E Rosser
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK.,Department of Neurology, Morriston Hospital, Swansea Bay University Health Board, Swansea SA6 6NL, UK.,School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| |
Collapse
|
57
|
Kim C, Yousefian-Jazi A, Choi SH, Chang I, Lee J, Ryu H. Non-Cell Autonomous and Epigenetic Mechanisms of Huntington's Disease. Int J Mol Sci 2021; 22:12499. [PMID: 34830381 PMCID: PMC8617801 DOI: 10.3390/ijms222212499] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
Huntington's disease (HD) is a rare neurodegenerative disorder caused by an expansion of CAG trinucleotide repeat located in the exon 1 of Huntingtin (HTT) gene in human chromosome 4. The HTT protein is ubiquitously expressed in the brain. Specifically, mutant HTT (mHTT) protein-mediated toxicity leads to a dramatic degeneration of the striatum among many regions of the brain. HD symptoms exhibit a major involuntary movement followed by cognitive and psychiatric dysfunctions. In this review, we address the conventional role of wild type HTT (wtHTT) and how mHTT protein disrupts the function of medium spiny neurons (MSNs). We also discuss how mHTT modulates epigenetic modifications and transcriptional pathways in MSNs. In addition, we define how non-cell autonomous pathways lead to damage and death of MSNs under HD pathological conditions. Lastly, we overview therapeutic approaches for HD. Together, understanding of precise neuropathological mechanisms of HD may improve therapeutic approaches to treat the onset and progression of HD.
Collapse
Affiliation(s)
- Chaebin Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (C.K.); (A.Y.-J.); (S.-H.C.)
| | - Ali Yousefian-Jazi
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (C.K.); (A.Y.-J.); (S.-H.C.)
| | - Seung-Hye Choi
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (C.K.); (A.Y.-J.); (S.-H.C.)
| | - Inyoung Chang
- Department of Biology, Boston University, Boston, MA 02215, USA;
| | - Junghee Lee
- Boston University Alzheimer’s Disease Research Center, Boston University, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, Boston, MA 02130, USA
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (C.K.); (A.Y.-J.); (S.-H.C.)
| |
Collapse
|
58
|
Kinnunen KM, Schwarz AJ, Turner EC, Pustina D, Gantman EC, Gordon MF, Joules R, Mullin AP, Scahill RI, Georgiou-Karistianis N. Volumetric MRI-Based Biomarkers in Huntington's Disease: An Evidentiary Review. Front Neurol 2021; 12:712555. [PMID: 34621236 PMCID: PMC8490802 DOI: 10.3389/fneur.2021.712555] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/10/2021] [Indexed: 01/02/2023] Open
Abstract
Huntington's disease (HD) is an autosomal-dominant inherited neurodegenerative disorder that is caused by expansion of a CAG-repeat tract in the huntingtin gene and characterized by motor impairment, cognitive decline, and neuropsychiatric disturbances. Neuropathological studies show that disease progression follows a characteristic pattern of brain atrophy, beginning in the basal ganglia structures. The HD Regulatory Science Consortium (HD-RSC) brings together diverse stakeholders in the HD community—biopharmaceutical industry, academia, nonprofit, and patient advocacy organizations—to define and address regulatory needs to accelerate HD therapeutic development. Here, the Biomarker Working Group of the HD-RSC summarizes the cross-sectional evidence indicating that regional brain volumes, as measured by volumetric magnetic resonance imaging, are reduced in HD and are correlated with disease characteristics. We also evaluate the relationship between imaging measures and clinical change, their longitudinal change characteristics, and within-individual longitudinal associations of imaging with disease progression. This analysis will be valuable in assessing pharmacodynamics in clinical trials and supporting clinical outcome assessments to evaluate treatment effects on neurodegeneration.
Collapse
Affiliation(s)
| | - Adam J Schwarz
- Takeda Pharmaceuticals, Ltd., Cambridge, MA, United States
| | | | - Dorian Pustina
- CHDI Management/CHDI Foundation, Princeton, NJ, United States
| | - Emily C Gantman
- CHDI Management/CHDI Foundation, Princeton, NJ, United States
| | - Mark F Gordon
- Teva Pharmaceuticals, West Chester, PA, United States
| | | | - Ariana P Mullin
- Critical Path Institute, Tucson, AZ, United States.,Wave Life Sciences, Ltd., Cambridge, MA, United States
| | - Rachael I Scahill
- Huntington's Disease Research Centre, UCL Institute of Neurology, London, United Kingdom
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| | | |
Collapse
|
59
|
Horta-Barba A, Martinez-Horta S, Perez-Perez J, Sampedro F, de Lucia N, De Michele G, Salvatore E, Kehrer S, Priller J, Migliore S, Squitieri F, Castaldo A, Mariotti C, Mañanes V, Lopez-Sendon JL, Rodriguez N, Martinez-Descals A, Júlio F, Janurio C, Delussi M, de Tommaso M, Noguera S, Ruiz-Idiago J, Sitek EJ, Wallner R, Nuzzi A, Pagonabarraga J, Kulisevsky J. Arithmetic Word-Problem Solving as Cognitive Marker of Progression in Pre-Manifest and Manifest Huntington's Disease. J Huntingtons Dis 2021; 10:459-468. [PMID: 34602494 DOI: 10.3233/jhd-210480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Arithmetic word-problem solving depends on the interaction of several cognitive processes that may be affected early in the disease in gene-mutation carriers for Huntington's disease (HD). OBJECTIVE Our goal was to examine the pattern of performance of arithmetic tasks in premanifest and manifest HD, and to examine correlations between arithmetic task performance and other neuropsychological tasks. METHODS We collected data from a multicenter cohort of 165 HD gene-mutation carriers. The sample consisted of 31 premanifest participants: 16 far-from (>12 years estimated time to diagnosis; preHD-A) and 15 close-to (≤12 years estimated time to diagnosis; preHD-B), 134 symptomatic patients (early-mild HD), and 37 healthy controls (HC). We compared performance between groups and explored the associations between arithmetic word-problem solving and neuropsychological and clinical variables. RESULTS Total arithmetic word-problem solving scores were lower in preHD-B patients than in preHD-A (p < 0.05) patients and HC (p < 0.01). Early-mild HD patients had lower scores than preHD patients (p < 0.001) and HC (p < 0.001). Compared to HC, preHD and early-mild HD participants made more errors as trial complexity increased. Moreover, arithmetic word-problem solving scores were significantly associated with measures of global cognition (p < 0.001), frontal-executive functions (p < 0.001), attention (p < 0.001) visual working memory (p < 0.001), mental rotation (p < 0.001), and confrontation naming (p < 0.05). CONCLUSION Arithmetic word-problem solving is affected early in the course of HD and is related to deficient processes in frontal-executive and mentalizing-related processes.
Collapse
Affiliation(s)
- Andrea Horta-Barba
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Barcelona, Spain.,European Huntington's Disease Network (EHDN)
| | - Saul Martinez-Horta
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Barcelona, Spain.,European Huntington's Disease Network (EHDN)
| | - Jesús Perez-Perez
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Barcelona, Spain.,European Huntington's Disease Network (EHDN)
| | - Frederic Sampedro
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Natascia de Lucia
- European Huntington's Disease Network (EHDN).,University of Naples "Federico II", Naples, Italy
| | - Giuseppe De Michele
- European Huntington's Disease Network (EHDN).,University of Naples "Federico II", Naples, Italy
| | - Elena Salvatore
- European Huntington's Disease Network (EHDN).,University of Naples "Federico II", Naples, Italy
| | - Stefanie Kehrer
- European Huntington's Disease Network (EHDN).,Department of Neuropsychiatry, Charité - Universitätsmedizin, Berlin, Germany
| | - Josef Priller
- European Huntington's Disease Network (EHDN).,Department of Neuropsychiatry, Charité - Universitätsmedizin, Berlin, Germany
| | - Simone Migliore
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo della, Sofferenza Research Hospital, San Giovanni Rotondo, Italy
| | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo della, Sofferenza Research Hospital, San Giovanni Rotondo, Italy
| | - Anna Castaldo
- European Huntington's Disease Network (EHDN).,Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Caterina Mariotti
- European Huntington's Disease Network (EHDN).,Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Veronica Mañanes
- European Huntington's Disease Network (EHDN).,Department of Neurology, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Jose Luis Lopez-Sendon
- European Huntington's Disease Network (EHDN).,Department of Neurology, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Noelia Rodriguez
- European Huntington's Disease Network (EHDN).,Department of Neurology. Fundación Jimenez Diaz, Madrid, Spain
| | - Asunción Martinez-Descals
- European Huntington's Disease Network (EHDN).,Department of Neurology. Fundación Jimenez Diaz, Madrid, Spain
| | - Filipa Júlio
- European Huntington's Disease Network (EHDN).,Coimbra Institute for Biomedical Imaging and Translational Research - CIBIT, University of Coimbra, Coimbra, Portugal.,Neurology Department, Coimbra University Hospital, Coimbra, Portugal
| | - Cristina Janurio
- European Huntington's Disease Network (EHDN).,Coimbra Institute for Biomedical Imaging and Translational Research - CIBIT, University of Coimbra, Coimbra, Portugal.,Neurology Department, Coimbra University Hospital, Coimbra, Portugal
| | - Marianna Delussi
- European Huntington's Disease Network (EHDN).,Applied Neurophysiology and Pain Unit, Apulian Center for Huntington's Disease SMBNOS Department, "Aldo Moro" University, Bari, Italy
| | - Marina de Tommaso
- European Huntington's Disease Network (EHDN).,Applied Neurophysiology and Pain Unit, Apulian Center for Huntington's Disease SMBNOS Department, "Aldo Moro" University, Bari, Italy
| | - Sandra Noguera
- European Huntington's Disease Network (EHDN).,Hospital Mare de Deu de la Mercè, Barcelona, Spain
| | - Jesus Ruiz-Idiago
- European Huntington's Disease Network (EHDN).,Hospital Mare de Deu de la Mercè, Barcelona, Spain
| | - Emilia J Sitek
- European Huntington's Disease Network (EHDN).,Department of Neurological and Psychiatric Nursing, Faculty of Health Science, Medical University of Gdansk, Gdańsk, Poland.,Department of Neurology, St. Adalbert Hospital, Gdańsk, Poland
| | - Renata Wallner
- Department of Psychiatry, Medical University of Wroclaw, Wroclaw, Poland
| | | | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Barcelona, Spain.,European Huntington's Disease Network (EHDN)
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Barcelona, Spain.,European Huntington's Disease Network (EHDN)
| | | |
Collapse
|
60
|
Lunven M, Hamet Bagnou J, Youssov K, Gabadinho A, Fliss R, Montillot J, Audureau E, Bapst B, Morgado G, Reilmann R, Schubert R, Busse M, Craufurd D, Massart R, Rosser A, Bachoud-Lévi AC. Cognitive decline in Huntington's disease in the Digitalized Arithmetic Task (DAT). PLoS One 2021; 16:e0253064. [PMID: 34424902 PMCID: PMC8382187 DOI: 10.1371/journal.pone.0253064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 05/25/2021] [Indexed: 11/20/2022] Open
Abstract
Background Efficient cognitive tasks sensitive to longitudinal deterioration in small cohorts of Huntington’s disease (HD) patients are lacking in HD research. We thus developed and assessed the digitized arithmetic task (DAT), which combines inner language and executive functions in approximately 4 minutes. Methods We assessed the psychometric properties of DAT in three languages, across four European sites, in 77 early-stage HD patients (age: 52 ± 11 years; 27 females), and 57 controls (age: 50 ± 10, 31 females). Forty-eight HD patients and 34 controls were followed up to one year with 96 participants who underwent MRI brain imaging (HD patients = 46) at baseline and 50 participants (HD patients = 22) at one year. Linear mixed models and Pearson correlations were used to assess associations with clinical assessment. Results At baseline, HD patients were less accurate (p = 0.0002) with increased response time (p<0.0001) when compared to DAT in controls. Test-retest reliability in HD patients ranged from good to excellent for response time (range: 0.63–0.79) and from questionable to acceptable for accuracy (range: r = 0.52–0.69). Only DAT, the Mattis Dementia Rating Scale, the Symbol Digit Modalities Test, and Total Functional Capacity scores were able to detect a decline within a one-year follow-up in HD patients (all p< 0.05). In contrast with all the other cognitive tasks, DAT correlated with striatal atrophy over time (p = 0.037) but not with motor impairment. Conclusions DAT is fast, reliable, motor-free, applicable in several languages, and able to unmask cognitive decline correlated with striatal atrophy in small cohorts of HD patients. This likely makes it a useful endpoint in future trials for HD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Marine Lunven
- Département d’Etudes Cognitives, École normale supérieure, PSL University, Paris, France
- University Paris Est Creteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, Creteil, France
- AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, Créteil, France
- NeurATRIS, Créteil, France
| | - Jennifer Hamet Bagnou
- Département d’Etudes Cognitives, École normale supérieure, PSL University, Paris, France
- University Paris Est Creteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, Creteil, France
- AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, Créteil, France
- NeurATRIS, Créteil, France
| | - Katia Youssov
- Département d’Etudes Cognitives, École normale supérieure, PSL University, Paris, France
- University Paris Est Creteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, Creteil, France
- AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, Créteil, France
- NeurATRIS, Créteil, France
| | - Alexis Gabadinho
- Département d’Etudes Cognitives, École normale supérieure, PSL University, Paris, France
- University Paris Est Creteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, Creteil, France
- AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, Créteil, France
- NeurATRIS, Créteil, France
| | - Rafika Fliss
- Département d’Etudes Cognitives, École normale supérieure, PSL University, Paris, France
- University Paris Est Creteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, Creteil, France
- AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, Créteil, France
- NeurATRIS, Créteil, France
| | - Justine Montillot
- Département d’Etudes Cognitives, École normale supérieure, PSL University, Paris, France
- University Paris Est Creteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, Creteil, France
- AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, Créteil, France
- NeurATRIS, Créteil, France
| | - Etienne Audureau
- Clinical Epidemiology and Ageing, Service de santé publique, Henri Mondor Hospital, AP-HP, Créteil, France
| | - Blanche Bapst
- Department of Neuroradiology, AP-HP, Henri Mondor University Hospital, Créteil, France
- Faculty of Medicine, Université Paris Est Créteil, Créteil, France
| | - Graça Morgado
- Centre d’Investigation Clinique, Hôpital Henri Mondor, Créteil, France
| | - Ralf Reilmann
- George-Huntington-Institute, Technology-Park, Muenster, Germany
- Department of Clinical Radiology University of Muenster, Muenster, Germany
- Dept. of Neurodegeneration and Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Robin Schubert
- George-Huntington-Institute, Technology-Park, Muenster, Germany
| | - Monica Busse
- Centre for Trials Research, Cardiff University, United Kingdom
- NMHRI, School of Medicine, and Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - David Craufurd
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Renaud Massart
- Département d’Etudes Cognitives, École normale supérieure, PSL University, Paris, France
- University Paris Est Creteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, Creteil, France
- AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, Créteil, France
| | - Anne Rosser
- NMHRI, School of Medicine, and Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
- Wales Brain Research And Intracranial Neurotherapeutics (BRAIN) unit, Wales
| | - Anne-Catherine Bachoud-Lévi
- Département d’Etudes Cognitives, École normale supérieure, PSL University, Paris, France
- University Paris Est Creteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, Creteil, France
- AP-HP, Hôpital Henri Mondor-Albert Chenevier, Centre de référence Maladie de Huntington, Service de Neurologie, Créteil, France
- NeurATRIS, Créteil, France
- * E-mail:
| |
Collapse
|
61
|
Rodrigues FB, Byrne LM, Lowe AJ, Tortelli R, Heins M, Flik G, Johnson EB, De Vita E, Scahill RI, Giorgini F, Wild EJ. Kynurenine pathway metabolites in cerebrospinal fluid and blood as potential biomarkers in Huntington's disease. J Neurochem 2021; 158:539-553. [PMID: 33797782 PMCID: PMC8375100 DOI: 10.1111/jnc.15360] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/18/2021] [Accepted: 03/26/2021] [Indexed: 01/31/2023]
Abstract
Converging lines of evidence from several models, and post-mortem human brain tissue studies, support the involvement of the kynurenine pathway (KP) in Huntington's disease (HD) pathogenesis. Quantifying KP metabolites in HD biofluids is desirable, both to study pathobiology and as a potential source of biomarkers to quantify pathway dysfunction and evaluate the biochemical impact of therapeutic interventions targeting its components. In a prospective single-site controlled cohort study with standardised collection of cerebrospinal fluid (CSF), blood, phenotypic and imaging data, we used high-performance liquid-chromatography to measure the levels of KP metabolites-tryptophan, kynurenine, kynurenic acid, 3-hydroxykynurenine, anthranilic acid and quinolinic acid-in CSF and plasma of 80 participants (20 healthy controls, 20 premanifest HD and 40 manifest HD). We investigated short-term stability, intergroup differences, associations with clinical and imaging measures and derived sample-size calculation for future studies. Overall, KP metabolites in CSF and plasma were stable over 6 weeks, displayed no significant group differences and were not associated with clinical or imaging measures. We conclude that the studied metabolites are readily and reliably quantifiable in both biofluids in controls and HD gene expansion carriers. However, we found little evidence to support a substantial derangement of the KP in HD, at least to the extent that it is reflected by the levels of the metabolites in patient-derived biofluids.
Collapse
Affiliation(s)
- Filipe B. Rodrigues
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Lauren M. Byrne
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Alexander J. Lowe
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Rosanna Tortelli
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | | | - Gunnar Flik
- Charles River LaboratoriesGroningenThe Netherlands
| | - Eileanoir B. Johnson
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Enrico De Vita
- Lysholm Department of NeuroradiologyNational Hospital for Neurology & NeurosurgeryLondonUK
- Department of Biomedical EngineeringSchool of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | - Rachael I. Scahill
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Flaviano Giorgini
- Department of Genetics and Genome BiologyUniversity of LeicesterLeicesterUK
| | - Edward J. Wild
- UCL Huntington's Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| |
Collapse
|
62
|
Migliore S, D'Aurizio G, Maffi S, Ceccarelli C, Ristori G, Romano S, Castaldo A, Mariotti C, Curcio G, Squitieri F. Cognitive and behavioral associated changes in manifest Huntington disease: A retrospective cross-sectional study. Brain Behav 2021; 11:e02151. [PMID: 34110097 PMCID: PMC8323039 DOI: 10.1002/brb3.2151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Behavioral and cognitive changes can be observed across all Huntington disease (HD) stages. Our multicenter and retrospective study investigated the association between cognitive and behavioral scale scores in manifest HD, at three different yearly timepoints. METHODS We analyzed cognitive and behavioral domains by the Unified Huntington's Disease Rating Scale (UHDRS) and by the Problem Behaviors Assessment Short Form (PBA-s), at three different yearly times of life (t0 or baseline, t1 after one year, t2 after two years), in 97 patients with manifest HD (mean age 48.62 ± 13.1), from three ENROLL-HD Centers. In order to test the disease progression, we also examined patients' motor and functional changes by the UHDRS, overtime. RESULTS The severity of apathy and of perseveration/obsession was associated with the severity of the cognitive decline (p < .0001), regardless of the yearly timepoint. The score of irritability significantly and positively correlated with perseveration errors in the verbal fluency test at t0 (r = .34; p = .001), while the psychosis significantly and negatively correlated with the information processing speed at t0 (r = -.21; p = .038) and significantly and positively correlated with perseveration errors in the verbal fluency test at t1 (r = .35; p < .0001). The disease progression was confirmed by the significant worsening of the UHDRS-Total Motor Score (TMS) and of the UHDRS-Total Functional Capacity (TFC) scale score after two-year follow-up (p < .0001). CONCLUSION Although the progression of abnormal behavioral manifestations cannot be predicted in HD, the severity of apathy and perseveration/obsessions are significantly associated with the severity of the cognitive function impairment, thus contributing, together, to the disease development and to patients' loss of independence, in addition to the neurological manifestations. This cognitive-behavior pattern determines a common underlying deficit depending on a dysexecutive syndrome.
Collapse
Affiliation(s)
- Simone Migliore
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| | - Giulia D'Aurizio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Sabrina Maffi
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| | - Consuelo Ceccarelli
- Italian League for Research on Huntington and Related Diseases (LIRH) Foundation, Rome, Italy
| | - Giovanni Ristori
- Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Centre for Experimental Neurological Therapies, S. Andrea Hospital, Sapienza University, Rome, Italy
| | - Silvia Romano
- Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Centre for Experimental Neurological Therapies, S. Andrea Hospital, Sapienza University, Rome, Italy
| | - Anna Castaldo
- Department of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Caterina Mariotti
- Department of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Curcio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, Fondazione IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| |
Collapse
|
63
|
Parkin GM, Corey-Bloom J, Snell C, Castleton J, Thomas EA. Plasma neurofilament light in Huntington's disease: A marker for disease onset, but not symptom progression. Parkinsonism Relat Disord 2021; 87:32-38. [PMID: 33940564 PMCID: PMC9083556 DOI: 10.1016/j.parkreldis.2021.04.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To investigate whether plasma NfL levels correlate with clinical symptom severity in premanifest (PM) and manifest HD (HD) individuals, and whether a NfL cut-point could distinguish PM from HD patients with reasonable accuracy. METHOD 98 participants (33 control, 26 PM, 39 HD), underwent blood sample collection and clinical assessment, using both UHDRS and non-UHDRS measures, at one academic HD Center. Years to onset (YTO), probability of disease onset in 5 years, and predicted years until 60% onset probability were also calculated. NfL levels were measured using a Meso Scale Discovery assay. RESULTS Cohorts differed by age. NfL levels differed significantly across diagnostic groups and were significantly correlated with age. Age-adjusted NfL levels were not correlated with clinical measures in either HD or PM cohorts, but were correlated when cohorts were combined. In PM subjects, NfL levels correlated with YTO, probability of onset in 5 years, and years until 60% onset probability. Using ROC analysis, a NfL cut-point of <53.15 pg/ml distinguished HD from control; <74.84 pg/ml distinguished HD from PM. CONCLUSIONS These findings implicate plasma NfL as a peripheral prognostic marker for premanifest-HD. Notably, we show that significant correlations between NfL and clinical symptoms are detected only when PM + HD subjects are combined, but not within HD subjects alone. To date, prior studies have investigated the clinical usefulness of NfL exclusively in merged PM + HD cohorts. Our data suggests a biasing of these previous correlations, and hence potentially limited usefulness of plasma NfL in monitoring HD symptom progression, for example, in clinical trials.
Collapse
Affiliation(s)
- Georgia M Parkin
- Department of Epidemiology, University of California Irvine, Irvine, CA, USA; Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA.
| | - Jody Corey-Bloom
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA.
| | - Chase Snell
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA.
| | - Jordan Castleton
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA.
| | - Elizabeth A Thomas
- Department of Epidemiology, University of California Irvine, Irvine, CA, USA; Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
64
|
Ghazaleh N, Houghton R, Palermo G, Schobel SA, Wijeratne PA, Long JD. Ranking the Predictive Power of Clinical and Biological Features Associated With Disease Progression in Huntington's Disease. Front Neurol 2021; 12:678484. [PMID: 34093422 PMCID: PMC8176643 DOI: 10.3389/fneur.2021.678484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/26/2021] [Indexed: 12/01/2022] Open
Abstract
Huntington's disease (HD) is characterised by a triad of cognitive, behavioural, and motor symptoms which lead to functional decline and loss of independence. With potential disease-modifying therapies in development, there is interest in accurately measuring HD progression and characterising prognostic variables to improve efficiency of clinical trials. Using the large, prospective Enroll-HD cohort, we investigated the relative contribution and ranking of potential prognostic variables in patients with manifest HD. A random forest regression model was trained to predict change of clinical outcomes based on the variables, which were ranked based on their contribution to the prediction. The highest-ranked variables included novel predictors of progression—being accompanied at clinical visit, cognitive impairment, age at diagnosis and tetrabenazine or antipsychotics use—in addition to established predictors, cytosine adenine guanine (CAG) repeat length and CAG-age product. The novel prognostic variables improved the ability of the model to predict clinical outcomes and may be candidates for statistical control in HD clinical studies.
Collapse
Affiliation(s)
| | | | | | | | - Peter A Wijeratne
- Department of Computer Science, Centre for Medical Imaging Computing, University College London, London, United Kingdom.,Department of Neurodegenerative Disease, Huntington's Disease Research Centre, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Jeffrey D Long
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Biostatistics, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
65
|
Schwarz AJ. The Use, Standardization, and Interpretation of Brain Imaging Data in Clinical Trials of Neurodegenerative Disorders. Neurotherapeutics 2021; 18:686-708. [PMID: 33846962 PMCID: PMC8423963 DOI: 10.1007/s13311-021-01027-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Imaging biomarkers play a wide-ranging role in clinical trials for neurological disorders. This includes selecting the appropriate trial participants, establishing target engagement and mechanism-related pharmacodynamic effect, monitoring safety, and providing evidence of disease modification. In the early stages of clinical drug development, evidence of target engagement and/or downstream pharmacodynamic effect-especially with a clear relationship to dose-can provide confidence that the therapeutic candidate should be advanced to larger and more expensive trials, and can inform the selection of the dose(s) to be further tested, i.e., to "de-risk" the drug development program. In these later-phase trials, evidence that the therapeutic candidate is altering disease-related biomarkers can provide important evidence that the clinical benefit of the compound (if observed) is grounded in meaningful biological changes. The interpretation of disease-related imaging markers, and comparability across different trials and imaging tools, is greatly improved when standardized outcome measures are defined. This standardization should not impinge on scientific advances in the imaging tools per se but provides a common language in which the results generated by these tools are expressed. PET markers of pathological protein aggregates and structural imaging of brain atrophy are common disease-related elements across many neurological disorders. However, PET tracers for pathologies beyond amyloid β and tau are needed, and the interpretability of structural imaging can be enhanced by some simple considerations to guard against the possible confound of pseudo-atrophy. Learnings from much-studied conditions such as Alzheimer's disease and multiple sclerosis will be beneficial as the field embraces rarer diseases.
Collapse
Affiliation(s)
- Adam J Schwarz
- Takeda Pharmaceuticals Ltd., 40 Landsdowne Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
66
|
Sampedro F, Pérez-Pérez J, Martínez-Horta S, Pérez-González R, Horta-Barba A, Campolongo A, Izquierdo C, Pagonabarraga J, Gómez-Ansón B, Kulisevsky J. Cortical microstructural correlates of plasma neurofilament light chain in Huntington's disease. Parkinsonism Relat Disord 2021; 85:91-94. [PMID: 33770670 DOI: 10.1016/j.parkreldis.2021.03.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/04/2021] [Accepted: 03/09/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Huntington's disease (HD) is a severe neurodegenerative disorder with no effective treatment. Minimally-invasive biomarkers such as blood neurofilament light chain (NfL) in HD are therefore needed to quantitatively characterize neuronal loss. NfL levels in HD are known to correlate with disease progression and striatal atrophy, but whether they also reflect cortical degeneration remains elusive. METHODS In a sample of 35 HD patients, we characterized the cortical macro (cortical thickness) and microstructural (increased intracortical diffusivity) correlates of plasma NfL levels. We further investigated whether NfL-related cortical alterations correlated with clinical indicators of disease progression. RESULTS Increased plasma NfL levels in HD reflected posterior-cortical microstructural degeneration, but not reduced cortical thickness (p < 0.05, corrected). Importantly, these imaging alterations correlated, in turn, with more severe motor, cognitive and behavioral symptoms. CONCLUSION Plasma NfL levels may be useful for tracking clinically-meaningful cortical deterioration in HD. Additionally, our results further reinforce the role of intracortical diffusivity as a valuable imaging indicator in movement disorders.
Collapse
Affiliation(s)
- Frederic Sampedro
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Jesus Pérez-Pérez
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Saul Martínez-Horta
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Rocío Pérez-González
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Andrea Horta-Barba
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Antonia Campolongo
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Cristina Izquierdo
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Beatriz Gómez-Ansón
- Neuroradiology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de La Santa Creu I Sant Pau, Barcelona, Spain; Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Spain.
| |
Collapse
|
67
|
Calderon-Villalon J, Ramirez-Garcia G, Fernandez-Ruiz J, Sangri-Gil F, Campos-Romo A, Galvez V. Planning deficits in Huntington's disease: A brain structural correlation by voxel-based morphometry. PLoS One 2021; 16:e0249144. [PMID: 33760890 PMCID: PMC7990304 DOI: 10.1371/journal.pone.0249144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/11/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Early Huntington’s disease (HD) patients begin to show planning deficits even before motor alterations start to manifest. Generally, planning ability is associated with the functioning of anterior brain areas such as the medial prefrontal cortex. However, early HD neuropathology involves significant atrophy in the occipital and parietal cortex, suggesting that more posterior regions could also be involved in these planning deficits. Objective To identify brain regions associated with planning deficits in HD patients at an early clinical stage. Materials and methods Twenty-two HD-subjects genetically confirmed with incipient clinical manifestation and twenty healthy subjects were recruited. All participants underwent MRI T1 image acquisition as well as testing in the Stockings of Cambridge (SOC) task to measure planning ability. First, group comparison of SOC measures were performed. Then, correlation voxel-based morphometry analyses were done between gray matter degeneration and SOC performance in the HD group. Results Accuracy and efficiency planning scores correlated with gray matter density in right lingual gyrus, middle temporal gyrus, anterior cingulate gyrus, and paracingulate gyrus. Conclusions Our results suggest that planning deficits exhibited by early HD-subjects are related to occipital and temporal cortical degeneration in addition to the frontal areas deterioration.
Collapse
Affiliation(s)
- Jesus Calderon-Villalon
- Laboratorio de Neurociencias Cognitivas y Desarrollo, Escuela de Psicología, Universidad Panamericana, Ciudad de México, México
| | - Gabriel Ramirez-Garcia
- Laboratorio de Neuropsicología, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Juan Fernandez-Ruiz
- Laboratorio de Neuropsicología, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
- Instituto de Neuroetología, Universidad Veracruzana, Ciudad de México, México
| | - Fernanda Sangri-Gil
- Laboratorio de Neurociencias Cognitivas y Desarrollo, Escuela de Psicología, Universidad Panamericana, Ciudad de México, México
| | - Aurelio Campos-Romo
- Unidad Periférica de Neurociencias, Facultad de Medicina, Instituto Nacional de Neurología y Neurocirugía “MVS”, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Victor Galvez
- Laboratorio de Neurociencias Cognitivas y Desarrollo, Escuela de Psicología, Universidad Panamericana, Ciudad de México, México
- Unidad Periférica de Neurociencias, Facultad de Medicina, Instituto Nacional de Neurología y Neurocirugía “MVS”, Universidad Nacional Autónoma de México, Ciudad de México, México
- * E-mail:
| |
Collapse
|
68
|
Woodgate S, Morgan-Jones P, Clinch S, Drew C, Playle R, Bennasar M, Hicks Y, Holt C, Bachoud-Lévi AC, Massart R, Craufurd D, Kirby N, Hamana K, Schubert R, Reilmann R, Rosser A, Busse M. Objectively characterizing Huntington's disease using a novel upper limb dexterity test. J Neurol 2021; 268:2550-2559. [PMID: 33555419 PMCID: PMC7868671 DOI: 10.1007/s00415-020-10375-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND The Clinch Token Transfer Test (C3t) is a bi-manual coin transfer task that incorporates cognitive tasks to add complexity. This study explored the concurrent and convergent validity of the C3t as a simple, objective assessment of impairment that is reflective of disease severity in Huntington's, that is not reliant on clinical expertise for administration. METHODS One-hundred-and-five participants presenting with pre-manifest (n = 16) or manifest (TFC-Stage-1 n = 39; TFC-Stage-2 n = 43; TFC-Stage-3 n = 7) Huntington's disease completed the Unified Huntington's Disease Rating Scale and the C3t at baseline. Of these, thirty-three were followed up after 12 months. Regression was used to estimate baseline individual and composite clinical scores (including cognitive, motor, and functional ability) using baseline C3t scores. Correlations between C3t and clinical scores were assessed using Spearman's R and visually inspected in relation to disease severity using scatterplots. Effect size over 12 months provided an indication of longitudinal behaviour of the C3t in relation to clinical measures. RESULTS Baseline C3t scores predicted baseline clinical scores to within 9-13% accuracy, being associated with individual and composite clinical scores. Changes in C3t scores over 12 months were small ([Formula: see text] ≤ 0.15) and mirrored the change in clinical scores. CONCLUSION The C3t demonstrates promise as a simple, easy to administer, objective outcome measure capable of predicting impairment that is reflective of Huntington's disease severity and offers a viable solution to support remote clinical monitoring. It may also offer utility as a screening tool for recruitment to clinical trials given preliminary indications of association with the prognostic index normed for Huntington's disease.
Collapse
Affiliation(s)
- Samuel Woodgate
- Cardiff School of Engineering, Cardiff University, Cardiff, United Kingdom
- NMRI, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Philippa Morgan-Jones
- Cardiff School of Engineering, Cardiff University, Cardiff, United Kingdom
- Centre for Trials Research, Cardiff University, 4th Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS, United Kingdom
| | - Susanne Clinch
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Cheney Drew
- Centre for Trials Research, Cardiff University, 4th Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS, United Kingdom
| | - Rebecca Playle
- Centre for Trials Research, Cardiff University, 4th Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS, United Kingdom
| | - Mohamed Bennasar
- School of Computing and Communications, The Open University, Milton Keynes, United Kingdom
| | - Yulia Hicks
- Cardiff School of Engineering, Cardiff University, Cardiff, United Kingdom
| | - Catherine Holt
- Cardiff School of Engineering, Cardiff University, Cardiff, United Kingdom
| | - Anne-Catherine Bachoud-Lévi
- Assistance Publique -Hopitaux de Paris, National Centre of Reference for Huntington's Disease, Neurology Department Henri Mondor Hospital, Creteil, France
- INSERM U955 01, Institut Mondor de Recherche Biomédicale, UPEC, Créteil-Ecole Normale Supérieure, PSL, Paris, France
| | - Renaud Massart
- Assistance Publique -Hopitaux de Paris, National Centre of Reference for Huntington's Disease, Neurology Department Henri Mondor Hospital, Creteil, France
- INSERM U955 01, Institut Mondor de Recherche Biomédicale, UPEC, Créteil-Ecole Normale Supérieure, PSL, Paris, France
| | - David Craufurd
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Nigel Kirby
- Centre for Trials Research, Cardiff University, 4th Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS, United Kingdom
| | - Katy Hamana
- School of Healthcare Sciences, Cardiff University, Cardiff, United Kingdom
| | - Robin Schubert
- George-Huntington-Institute, Technology-Park, Muenster, Germany
| | - Ralf Reilmann
- George-Huntington-Institute, Technology-Park, Muenster, Germany
- Department of Clinical Radiology, University of Muenster, Muenster, Germany
- Department of Neurodegeneration and Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Anne Rosser
- NMRI, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Monica Busse
- Centre for Trials Research, Cardiff University, 4th Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS, United Kingdom.
| |
Collapse
|
69
|
Bachoud-Lévi AC, Massart R, Rosser A. Cell therapy in Huntington's disease: Taking stock of past studies to move the field forward. Stem Cells 2021; 39:144-155. [PMID: 33176057 PMCID: PMC10234449 DOI: 10.1002/stem.3300] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/01/2020] [Accepted: 10/20/2020] [Indexed: 06/02/2023]
Abstract
Huntington's disease (HD) is a rare inherited neurodegenerative disease that manifests mostly in adulthood with progressive cognitive, behavioral, and motor dysfunction. Neuronal loss occurs predominantly in the striatum but also extends to other brain regions, notably the cortex. Most patients die around 20 years after motor onset, although there is variability in the rate of progression and some phenotypic heterogeneity. The most advanced experimental therapies currently are huntingtin-lowering strategies, some of which are in stage 3 clinical trials. However, even if these approaches are successful, it is unlikely that they will be applicable to all patients or will completely halt continued loss of neural cells in all cases. On the other hand, cellular therapies have the potential to restore atrophied tissues and may therefore provide an important complementary therapeutic avenue. Pilot studies of fetal cell grafts in the 2000s reported the most dramatic clinical improvements yet achieved for this disease, but subsequent studies have so far failed to identify methodology to reliably reproduce these results. Moving forward, a major challenge will be to generate suitable donor cells from (nonfetal) cell sources, but in parallel there are a host of procedural and trial design issues that will be important for improving reliability of transplants and so urgently need attention. Here, we consider findings that have emerged from clinical transplant studies in HD to date, in particular new findings emerging from the recent multicenter intracerebral transplant HD study, and consider how these data may be used to inform future cell therapy trials.
Collapse
Affiliation(s)
- Anne-Catherine Bachoud-Lévi
- Assistance Publique-Hôpitaux de Paris, National Reference Center for Huntington's Disease, Neurology Department, Henri Mondor-Albert Chenevier Hospital, Créteil, France
- Département d'Etudes Cognitives, École Normale Supérieure, PSL University, Paris, France
- Inserm U955, Institut Mondor de Recherche Biomédicale, Equipe E01 NeuroPsychologie Interventionnelle, Créteil, France
- NeurATRIS, Créteil, France
- Université Paris-Est Créteil, Faculté de Médecine, Créteil, France
| | - Renaud Massart
- Assistance Publique-Hôpitaux de Paris, National Reference Center for Huntington's Disease, Neurology Department, Henri Mondor-Albert Chenevier Hospital, Créteil, France
- Département d'Etudes Cognitives, École Normale Supérieure, PSL University, Paris, France
- Inserm U955, Institut Mondor de Recherche Biomédicale, Equipe E01 NeuroPsychologie Interventionnelle, Créteil, France
- NeurATRIS, Créteil, France
| | - Anne Rosser
- Centre for Trials Research, Cardiff University, Cardiff, UK
- Cardiff University Brain Repair Group, Life Sciences Building, School of Biosciences, Cardiff, UK
- Neuroscience and Mental Health Research Institute and Division of Psychological Medicine and Clinical Neurosciences, Hadyn Ellis Building, Cardiff, UK
- Brain Repair And Intracranial Neurotherapeutics (BRAIN) Unit, Cardiff University, Cardiff, UK
| |
Collapse
|
70
|
Griffin BA, Booth MS, Busse M, Wild EJ, Setodji C, Warner JH, Sampaio C, Mohan A. Estimating the causal effects of modifiable, non-genetic factors on Huntington disease progression using propensity score weighting. Parkinsonism Relat Disord 2021; 83:56-62. [PMID: 33476879 PMCID: PMC7949328 DOI: 10.1016/j.parkreldis.2021.01.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Despite being genetically inherited, it is unclear how non-genetic factors (e.g., substance use, employment) might contribute to the progression and severity of Huntington's disease (HD). METHODS We used propensity score (PS) weighting in a large (n = 2914) longitudinal dataset (Enroll-HD) to examine the impact of education, employment status, and use of tobacco, alcohol, and recreational and therapeutic drugs on HD progression. Each factor was investigated in isolation while controlling for 19 other factors to ensure that groups were balanced at baseline on potential confounders using PS weights. Outcomes were compared several years later using doubly robust models. RESULTS Our results highlighted cases where modifiable (non-genetic) factors - namely light and moderate alcohol use and employment - would have been associated with HD progression in models that did not use PS weights to control for baseline imbalances. These associations did not hold once we applied PS weights to balance baseline groups. We also found potential evidence of a protective effect of substance use (primarily marijuana use), and that those who needed antidepressant treatment were likely to progress faster than non-users. CONCLUSIONS Our study is the first to examine the effect of non-genetic factors on HD using a novel application of PS weighting. We show that previously-reported associated factors - including light and moderate alcohol use - are reduced and no longer significantly linked to HD progression after PS weighting. This indicates the potential value of PS weighting in examining non-genetic factors contributing to HD as well as in addressing the known biases that occur with observational data.
Collapse
Affiliation(s)
- Beth Ann Griffin
- RAND Center for Causal Inference, RAND Corporation, 1200, South Hayes Street, Arlington, VA, USA.
| | | | - Monica Busse
- Centre for Trials Research, Cardiff University, Neuadd Merionydd, Heath Park, CF14 4XN, Cardiff, UK
| | - Edward J Wild
- Huntington's Disease Centre, UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Claude Setodji
- RAND Center for Causal Inference, RAND Corporation, 4570, Fifth Ave #600, Pittsburgh, PA, USA
| | - John H Warner
- CHDI Management/CHDI Foundation, 155 Village Boulevard, Suite 200, Princeton, NJ, USA
| | - Cristina Sampaio
- CHDI Management/CHDI Foundation, 155 Village Boulevard, Suite 200, Princeton, NJ, USA
| | - Amrita Mohan
- CHDI Management/CHDI Foundation, 155 Village Boulevard, Suite 200, Princeton, NJ, USA
| |
Collapse
|
71
|
Estevez-Fraga C, Scahill RI, Durr A, Leavitt BR, Roos RAC, Langbehn DR, Rees G, Gregory S, Tabrizi SJ. Composite UHDRS Correlates With Progression of Imaging Biomarkers in Huntington's Disease. Mov Disord 2021; 36:1259-1264. [PMID: 33471951 DOI: 10.1002/mds.28489] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/10/2020] [Accepted: 12/07/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The composite Unified Huntington's Disease Rating Scale (cUHDRS) is a multidimensional measure of progression in Huntington's disease (HD) being used as a primary outcome in clinical trials investigating potentially disease-modifying huntingtin-lowering therapies. OBJECTIVE Evaluating volumetric and structural connectivity correlates of the cUHDRS. METHODS One hundred and nineteen premanifest and 119 early-HD participants were included. Gray and white matter (WM) volumes were correlated with cUHDRS cross-sectionally and longitudinally using voxel-based morphometry. Correlations between baseline fractional anisotropy (FA); mean, radial, and axial diffusivity; and baseline cUHDRS were examined using tract-based spatial statistics. RESULTS Worse performance in the cUHDRS over time correlated with longitudinal volume decreases in the occipito-parietal cortex and centrum semiovale, whereas lower baseline scores correlated with decreased volume in the basal ganglia and surrounding WM. Lower cUHDRS scores were also associated with reduced FA and increased diffusivity at baseline. CONCLUSION The cUHDRS correlates with imaging biomarkers and tracks atrophy progression in HD supporting its biological relevance. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Carlos Estevez-Fraga
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Rachael I Scahill
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Alexandra Durr
- Sorbonne Université, Paris Brain Institute (ICM), AP-HP, Inserm, CNRS, Pitié-Salpêtrière University Hospital, Paris, France
| | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Raymund A C Roos
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Geraint Rees
- Wellcome Centre for Neuroimaging, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sarah Gregory
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Sarah J Tabrizi
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
72
|
Lang C, Gries C, Lindenberg KS, Lewerenz J, Uhl S, Olsson C, Samzelius J, Landwehrmeyer GB. Monitoring the Motor Phenotype in Huntington's Disease by Analysis of Keyboard Typing During Real Life Computer Use. J Huntingtons Dis 2021; 10:259-268. [PMID: 33459656 DOI: 10.3233/jhd-200451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Besides cognitive and psychiatric abnormalities, motor symptoms are the most prominent in Huntington's disease. The manifest disease is preceded by a prodromal phase with subtle changes such as fine motor disturbances or concentration problems. OBJECTIVE Movement disorders show a high variation in their clinical manifestation depending on condition and external influences. Therefore, devices for continuous measurements, which patients use in their daily life and which can monitor motor abnormalities, in addition to the medical examination, might be useful. The aim of current scientific efforts is to find markers that reflect the prodromal phase in gene carriers. This is important for future interventional studies, as future therapies should be applied at the stage of neuronal dysfunction, i.e., before the clinical manifestation. METHODS We performed a software-supported, continuous monitoring of keyboard typing on the participants' own computer to evaluate this method as a tool to assess the motor phenotype in HD. We included 40 participants and obtained sufficient data from 25 participants, 12 of whom were manifest HD patients, 7 HD gene expansion carriers (HDGEC) and 6 healthy controls. RESULTS In a cross-sectional analysis we found statistically significant higher typing inconsistency in HD patients compared to controls. Typing inconsistency compared between HDGEC and healthy controls showed a trend to higher inconsistency levels in HDGEC. We found correlations between typing cadence and clinical scores: the UHDRS finger tapping item, the composite UHDRS and the CAP score. CONCLUSION The typing cadence inconsistency is an appropriate marker to evaluate fine motor skills of HD patients and HDGEC and is correlated to established clinical measurements.
Collapse
Affiliation(s)
- Christina Lang
- Department of Neurology, University Hospital Ulm, Ulm, Germany
| | | | | | - Jan Lewerenz
- Department of Neurology, University Hospital Ulm, Ulm, Germany
| | - Stefanie Uhl
- Department of Neurology, University Hospital Ulm, Ulm, Germany
| | | | | | | |
Collapse
|
73
|
Rodrigues FB, Byrne LM, Tortelli R, Johnson EB, Wijeratne PA, Arridge M, De Vita E, Ghazaleh N, Houghton R, Furby H, Alexander DC, Tabrizi SJ, Schobel S, Scahill RI, Heslegrave A, Zetterberg H, Wild EJ. Mutant huntingtin and neurofilament light have distinct longitudinal dynamics in Huntington's disease. Sci Transl Med 2020; 12:eabc2888. [PMID: 33328328 PMCID: PMC7611886 DOI: 10.1126/scitranslmed.abc2888] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 09/04/2020] [Indexed: 07/26/2023]
Abstract
The longitudinal dynamics of the most promising biofluid biomarker candidates for Huntington's disease (HD)-mutant huntingtin (mHTT) and neurofilament light (NfL)-are incompletely defined. Characterizing changes in these candidates during disease progression could increase our understanding of disease pathophysiology and help the identification of effective therapies. In an 80-participant cohort over 24 months, mHTT in cerebrospinal fluid (CSF), as well as NfL in CSF and blood, had distinct longitudinal trajectories in HD mutation carriers compared with controls. Baseline analyte values predicted clinical disease status, subsequent clinical progression, and brain atrophy, better than did the rate of change in analytes. Overall, NfL was a stronger monitoring and prognostic biomarker for HD than mHTT. Nonetheless, mHTT has prognostic value and might be a valuable pharmacodynamic marker for huntingtin-lowering trials.
Collapse
Affiliation(s)
- Filipe B Rodrigues
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London WC1B 5EH, UK
| | - Lauren M Byrne
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London WC1B 5EH, UK
| | - Rosanna Tortelli
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London WC1B 5EH, UK
| | - Eileanoir B Johnson
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London WC1B 5EH, UK
| | - Peter A Wijeratne
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1E 6BT, UK
| | - Marzena Arridge
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
- Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Enrico De Vita
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
- Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Naghmeh Ghazaleh
- PD Personalised Healthcare, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Richard Houghton
- PD Personalised Healthcare, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Hannah Furby
- PD Personalised Healthcare, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Daniel C Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London WC1E 6BT, UK
| | - Sarah J Tabrizi
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London WC1B 5EH, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Scott Schobel
- Product Development Neuroscience, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Rachael I Scahill
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London WC1B 5EH, UK
| | - Amanda Heslegrave
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 431 80 Mölndal, Sweden
| | - Edward J Wild
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London WC1B 5EH, UK.
| |
Collapse
|
74
|
Tan B, Shishegar R, Poudel GR, Fornito A, Georgiou-Karistianis N. Cortical morphometry and neural dysfunction in Huntington's disease: a review. Eur J Neurol 2020; 28:1406-1419. [PMID: 33210786 DOI: 10.1111/ene.14648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/22/2020] [Accepted: 11/12/2020] [Indexed: 01/09/2023]
Abstract
Numerous neuroimaging techniques have been used to identify biomarkers of disease progression in Huntington's disease (HD). To date, the earliest and most sensitive of these is caudate volume; however, it is becoming increasingly evident that numerous changes to cortical structures, and their interconnected networks, occur throughout the course of the disease. The mechanisms by which atrophy spreads from the caudate to these cortical regions remains unknown. In this review, the neuroimaging literature specific to T1-weighted and diffusion-weighted magnetic resonance imaging is summarized and new strategies for the investigation of cortical morphometry and the network spread of degeneration in HD are proposed. This new avenue of research may enable further characterization of disease pathology and could add to a suite of biomarker/s of disease progression for patient stratification that will help guide future clinical trials.
Collapse
Affiliation(s)
- Brendan Tan
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| | - Rosita Shishegar
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.,Australian e-Health Research Centre, CSIRO, Melbourne, VIC, Australia.,Monash Biomedical Imaging, Melbourne, VIC, Australia
| | - Govinda R Poudel
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.,Sydney Imaging, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Australian Catholic University, Melbourne, VIC, Australia
| | - Alex Fornito
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.,Monash Biomedical Imaging, Melbourne, VIC, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
75
|
Niemela V, Landtblom AM, Nyholm D, Kneider M, Constantinescu R, Paucar M, Svenningsson P, Abujrais S, Burman J, Shevchenko G, Bergquist J, Sundblom J. Proenkephalin Decreases in Cerebrospinal Fluid with Symptom Progression of Huntington's Disease. Mov Disord 2020; 36:481-491. [PMID: 33247616 PMCID: PMC7984171 DOI: 10.1002/mds.28391] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/07/2020] [Accepted: 10/21/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Identifying molecular changes that contribute to the onset and progression of Huntington's disease (HD) is of importance for the development and evaluation of potential therapies. METHODS We conducted an unbiased mass-spectrometry proteomic analysis on the cerebrospinal fluid of 12 manifest HD patients (ManHD), 13 pre-manifest (preHD), and 38 controls. A biologically plausible and significant possible biomarker was validated in samples from a separate cohort of patients and controls consisting of 23 ManHD patients and 23 controls. RESULTS In ManHD compared to preHD, 10 proteins were downregulated and 43 upregulated. Decreased levels of proenkephalin (PENK) and transthyretin were closely linked to HD symptom severity, whereas levels of 15 upregulated proteins were associated with symptom severity. The decreased PENK levels were replicated in the separate cohort where absolute quantitation was performed. CONCLUSIONS We hypothesize that declining PENK levels reflect the degeneration of medium spiny neurons (MSNs) that produce PENK and that assays for PENK may serve as a surrogate marker for the state of MSNs in HD. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Valter Niemela
- Department of Neuroscience; Neurology, Uppsala University, Uppsala, Sweden
| | | | - Dag Nyholm
- Department of Neuroscience; Neurology, Uppsala University, Uppsala, Sweden
| | - Maria Kneider
- Institute of Neuroscience and Physiology; Clinical Neuroscience, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Radu Constantinescu
- Institute of Neuroscience and Physiology; Clinical Neuroscience, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Martin Paucar
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sandy Abujrais
- Analytical Chemistry, Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Joachim Burman
- Department of Neuroscience; Neurology, Uppsala University, Uppsala, Sweden
| | - Ganna Shevchenko
- Analytical Chemistry, Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Jonas Bergquist
- Analytical Chemistry, Department of Chemistry - BMC, Uppsala University, Uppsala, Sweden
| | - Jimmy Sundblom
- Department of Neuroscience; Neurosurgery, Uppsala University, Uppsala, Sweden
| |
Collapse
|
76
|
Lowe AJ, Sjödin S, Rodrigues FB, Byrne LM, Blennow K, Tortelli R, Zetterberg H, Wild EJ. Cerebrospinal fluid endo-lysosomal proteins as potential biomarkers for Huntington's disease. PLoS One 2020; 15:e0233820. [PMID: 32804976 PMCID: PMC7430717 DOI: 10.1371/journal.pone.0233820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/30/2020] [Indexed: 01/13/2023] Open
Abstract
Molecular markers derived from cerebrospinal fluid (CSF) represent an accessible means of exploring the pathobiology of Huntington's disease (HD) in vivo. The endo-lysosomal/autophagy system is dysfunctional in HD, potentially contributing to disease pathogenesis and representing a potential target for therapeutic intervention. Several endo-lysosomal proteins have shown promise as biomarkers in other neurodegenerative diseases; however, they have yet to be fully explored in HD. We performed parallel reaction monitoring mass spectrometry analysis (PRM-MS) of multiple endo-lysosomal proteins in the CSF of 60 HD mutation carriers and 20 healthy controls. Using generalised linear models controlling for age and CAG, none of the 18 proteins measured displayed significant differences in concentration between HD patients and controls. This was affirmed by principal component analysis, in which no significant difference across disease stage was found in any of the three components representing lysosomal hydrolases, binding/transfer proteins and innate immune system/peripheral proteins. However, several proteins were associated with measures of disease severity and cognition: most notably amyloid precursor protein, which displayed strong correlations with composite Unified Huntington's Disease Rating Scale, UHDRS Total Functional Capacity, UHDRS Total Motor Score, Symbol Digit Modalities Test and Stroop Word Reading. We conclude that although endo-lysosomal proteins are unlikely to have value as disease state CSF biomarkers for Huntington's disease, several proteins demonstrate associations with clinical severity, thus warranting further, targeted exploration and validation in larger, longitudinal samples.
Collapse
Affiliation(s)
- Alexander J. Lowe
- UCL Huntington’s Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Simon Sjödin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Filipe B. Rodrigues
- UCL Huntington’s Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Lauren M. Byrne
- UCL Huntington’s Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Rosanna Tortelli
- UCL Huntington’s Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Henrik Zetterberg
- UCL Huntington’s Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Edward J. Wild
- UCL Huntington’s Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
77
|
Tabrizi SJ, Flower MD, Ross CA, Wild EJ. Huntington disease: new insights into molecular pathogenesis and therapeutic opportunities. Nat Rev Neurol 2020; 16:529-546. [PMID: 32796930 DOI: 10.1038/s41582-020-0389-4] [Citation(s) in RCA: 297] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2020] [Indexed: 12/11/2022]
Abstract
Huntington disease (HD) is a neurodegenerative disease caused by CAG repeat expansion in the huntingtin gene (HTT) and involves a complex web of pathogenic mechanisms. Mutant HTT (mHTT) disrupts transcription, interferes with immune and mitochondrial function, and is aberrantly modified post-translationally. Evidence suggests that the mHTT RNA is toxic, and at the DNA level, somatic CAG repeat expansion in vulnerable cells influences the disease course. Genome-wide association studies have identified DNA repair pathways as modifiers of somatic instability and disease course in HD and other repeat expansion diseases. In animal models of HD, nucleocytoplasmic transport is disrupted and its restoration is neuroprotective. Novel cerebrospinal fluid (CSF) and plasma biomarkers are among the earliest detectable changes in individuals with premanifest HD and have the sensitivity to detect therapeutic benefit. Therapeutically, the first human trial of an HTT-lowering antisense oligonucleotide successfully, and safely, reduced the CSF concentration of mHTT in individuals with HD. A larger trial, powered to detect clinical efficacy, is underway, along with trials of other HTT-lowering approaches. In this Review, we discuss new insights into the molecular pathogenesis of HD and future therapeutic strategies, including the modulation of DNA repair and targeting the DNA mutation itself.
Collapse
Affiliation(s)
- Sarah J Tabrizi
- Huntington's Disease Centre, University College London, London, UK. .,Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK. .,UK Dementia Research Institute, University College London, London, UK.
| | - Michael D Flower
- Huntington's Disease Centre, University College London, London, UK.,Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK.,UK Dementia Research Institute, University College London, London, UK
| | - Christopher A Ross
- Departments of Neurology, Neuroscience and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Edward J Wild
- Huntington's Disease Centre, University College London, London, UK.,Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
78
|
Pini L, Youssov K, Sambataro F, Bachoud‐Levi A, Vallesi A, Jacquemot C. Striatal connectivity in pre‐manifest Huntington’s disease is differentially affected by disease burden. Eur J Neurol 2020; 27:2147-2157. [DOI: 10.1111/ene.14423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/25/2020] [Indexed: 11/26/2022]
Affiliation(s)
- L. Pini
- Department of Neuroscience & Padova Neuroscience Center University of Padova Padova Italy
| | - K. Youssov
- Département d'Études Cognitives École Normale Supérieure PSL University ParisFrance
- Faculté de Santé Université Paris‐Est Créteil CréteilFrance
- Inserm U955 Equipe E01 NeuroPsychologie Interventionnelle Institut Mondor de Recherche Biomédicale CréteilFrance
- Centre de référence Maladie de Huntington Service de Neurologie Hôpital Henri Mondor, AP‐HP Créteil France
| | - F. Sambataro
- Department of Neuroscience & Padova Neuroscience Center University of Padova Padova Italy
| | - A.‐C. Bachoud‐Levi
- Département d'Études Cognitives École Normale Supérieure PSL University ParisFrance
- Faculté de Santé Université Paris‐Est Créteil CréteilFrance
- Inserm U955 Equipe E01 NeuroPsychologie Interventionnelle Institut Mondor de Recherche Biomédicale CréteilFrance
- Centre de référence Maladie de Huntington Service de Neurologie Hôpital Henri Mondor, AP‐HP Créteil France
| | - A. Vallesi
- Department of Neuroscience & Padova Neuroscience Center University of Padova Padova Italy
- Brain Imaging and Neural Dynamics Research Group IRCCS San Camillo Hospital Venice Italy
| | - C. Jacquemot
- Département d'Études Cognitives École Normale Supérieure PSL University ParisFrance
- Faculté de Santé Université Paris‐Est Créteil CréteilFrance
- Inserm U955 Equipe E01 NeuroPsychologie Interventionnelle Institut Mondor de Recherche Biomédicale CréteilFrance
| |
Collapse
|
79
|
Langbehn DR, Hersch S. Clinical Outcomes and Selection Criteria for Prodromal Huntington's Disease Trials. Mov Disord 2020; 35:2193-2200. [PMID: 32686867 PMCID: PMC7818458 DOI: 10.1002/mds.28222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/04/2020] [Accepted: 06/30/2020] [Indexed: 11/08/2022] Open
Abstract
Background Huntington's disease (HD) develops in individuals with extended cytosine‐adenine‐guanine (CAG) repeats within the huntingtin (HTT) gene, causing neurodegeneration and progressive motor and cognitive symptoms. The inclusion of mutant HTT carriers in whom overt symptoms are not yet fully manifest in therapeutic trials would enable the development of treatments that delay or halt the accumulation of significant disability. Objectives The present analyses assess whether screening prediagnosis (preHD) individuals based on a normalized prognostic index (PIN) score would enable the selection of prodromal preHD subjects in whom longitudinal changes in established outcome measures might provide robust signals. It also compares the relative statistical effect size of longitudinal change for these measures. Methods Individual participant data from 2 studies were used to develop mixed effect linear models to assess longitudinal changes in clinical metrics for participants with preHD and PIN‐stratified subcohorts. Relative effect sizes were calculated in 5 preHD studies and internally normalized to evaluate the strength and consistency of each metric across cohorts. Results Longitudinal modeling data demonstrate the amplification of effect sizes when preHD subcohorts were selected by PIN score thresholds of >0.0 and >0.4. These models and relative effect sizes across 5 studies consistently indicate that the Unified Huntington's Disease Rating Scale total motor score exhibits the greatest change in preHD. Conclusions These analyses suggest that the employment of PIN scores to homogenize and stratify preHD cohorts could improve the efficiency of current outcome measures, the most robust of which is the total motor score. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Douglas R Langbehn
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Steven Hersch
- Voyager Therapeutics, Inc., Cambridge, Massachusetts, USA.,Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
80
|
Bachoud-Lévi AC. Human Fetal Cell Therapy in Huntington's Disease: A Randomized, Multicenter, Phase II Trial. Mov Disord 2020; 35:1323-1335. [PMID: 32666599 DOI: 10.1002/mds.28201] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Huntington's disease is a rare, severe, inherited neurodegenerative disease in which we assessed the safety and efficacy of grafting human fetal ganglionic eminence intrastriatally. METHODS Patients at the early stage of the disease were enrolled in the Multicentric Intracerebral Grafting in Huntington's Disease trial, a delayed-start phase II randomized study. After a run-in period of 12 months, patients were randomized at month 12 to either the treatment group (transplanted at month 13-month 14) or the control group and secondarily treated 20 months later (month 33-month 34). The primary outcome was total motor score compared between both groups 20 months postrandomization (month 32). Secondary outcomes included clinical, imaging, and electrophysiological findings and a comparison of pregraft and postgraft total motor score slopes during the entire study period (month 0-month 52) regardless of the time of transplant. RESULTS Of 54 randomized patients, 45 were transplanted; 26 immediately (treatment) and 19 delayed (control). Mean total motor score at month 32 did not differ between groups (treated controls difference in means adjusted for M12: +2.9 [95% confidence interval, -2.8 to 8.6]; P = 0.31). Its rate of decline after transplantation was similar to that before transplantation. A total of 27 severe adverse events were recorded in the randomized patients, 10 of which were related to the transplant procedure. Improvement of procedures during the trial significantly decreased the frequency of surgical events.We found antihuman leucocytes antigen antibodies in 40% of the patients. CONCLUSION No clinical benefit was found in this trial. This may have been related to graft rejection. Ectopia and high track number negatively influence the graft outcome. Procedural adjustments substantially improved surgical safety. (ClinicalTrials.gov NCT00190450.) © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Anne-Catherine Bachoud-Lévi
- Assistance Publique-Hôpitaux de Paris, National Reference Center for Huntington's Disease, Neurology Department, Henri Mondor-Albert Chenevier Hospital, Créteil, France.,Equipe neuropsychologie interventionnelle, Département d'études cognitives, École normale supérieure, PSL, Research University, Institut Mondor de Recherche biomédicale, Université Paris-Est, INSERM, Paris, and Créteil, France.,Faculté de Santé, Université Paris Est, Créteil, France
| | | |
Collapse
|
81
|
Steventon JJ, Furby H, Ralph J, O'Callaghan P, Rosser AE, Wise RG, Busse M, Murphy K. Altered cerebrovascular response to acute exercise in patients with Huntington's disease. Brain Commun 2020; 2:fcaa044. [PMID: 32566927 PMCID: PMC7293798 DOI: 10.1093/braincomms/fcaa044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/26/2020] [Accepted: 03/16/2020] [Indexed: 01/20/2023] Open
Abstract
The objective of this study was to determine whether a single session of exercise was sufficient to induce cerebral adaptations in individuals with Huntington’s disease and to explore the time dynamics of any acute cerebrovascular response. In this case–control study, we employed arterial-spin labelling MRI in 19 Huntington’s disease gene-positive participants (32–65 years, 13 males) and 19 controls (29–63 years, 10 males) matched for age, gender, body mass index and self-reported activity levels, to measure global and regional perfusion in response to 20 min of moderate-intensity cycling. Cerebral perfusion was measured at baseline and 15, 40 and 60 min after exercise cessation. Relative to baseline, we found that cerebral perfusion increased in patients with Huntington’s disease yet was unchanged in control participants in the precentral gyrus (P = 0.016), middle frontal gyrus (P = 0.046) and hippocampus (P = 0.048) 40 min after exercise cessation (+15 to +32.5% change in Huntington’s disease participants, −7.7 to 0.8% change in controls). The length of the disease‐causing trinucleotide repeat expansion in the huntingtin gene predicted the change in the precentral gyrus (P = 0.03) and the intensity of the exercise intervention predicted hippocampal perfusion change in Huntington’s disease participants (P < 0.001). In both groups, exercise increased hippocampal blood flow 60 min after exercise cessation (P = 0.039). These findings demonstrate the utility of acute exercise as a clinically sensitive experimental paradigm to modulate the cerebrovasculature. Twenty minutes of aerobic exercise induced transient cerebrovascular adaptations in the hippocampus and cortex selectively in Huntington’s disease participants and likely represents latent neuropathology not evident at rest.
Collapse
Affiliation(s)
- Jessica J Steventon
- Cardiff University Brain Research Imaging Centre, School of Physics and Astronomy, Cardiff University, Cardiff CF24 4HQ, UK.,Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff CF24 4HQ, UK
| | - Hannah Furby
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff CF24 4HQ, UK.,Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff CF24 4HQ, UK
| | - James Ralph
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff CF24 4HQ, UK
| | - Peter O'Callaghan
- Cardiology Department, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Anne E Rosser
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff CF24 4HQ, UK.,Cardiff Brain Repair Group, School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Richard G Wise
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff CF24 4HQ, UK
| | - Monica Busse
- Centre for Trials Research, Cardiff University, Cardiff CF14 4YS, UK
| | - Kevin Murphy
- Cardiff University Brain Research Imaging Centre, School of Physics and Astronomy, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
82
|
Mills JA, Long JD, Mohan A, Ware JJ, Sampaio C. Cognitive and Motor Norms for Huntington’s Disease. Arch Clin Neuropsychol 2020; 35:671-682. [DOI: 10.1093/arclin/acaa026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/21/2020] [Accepted: 03/20/2020] [Indexed: 11/12/2022] Open
Abstract
Abstract
Background
The progression of Huntington’s disease (HD) for gene-expanded carriers is well-studied. Natural aging effects, however, are not often considered in the evaluation of HD progression.
Objective
To examine the effects of natural aging for healthy controls and to develop normative curves by age, sex, and education from the distribution of observed scores for the Symbol Digit Modalities Test, Stroop Word Reading Test, Stroop Color Naming Test, Stroop Interference Test, Total Motor Score, and Total Functional Capacity (TFC) from the Unified Huntington’s Disease Rating Scale (UHDRS) along with a composite score.
Methods
After combining longitudinal REGISTRY and Enroll-HD data, we used quantile regression and natural cubic splines for age to fit models for healthy controls (N = 3,394; N observations = 8,619). Normative curves were estimated for the 0.05, 0.25, 0.50, 0.75, and 0.95 quantiles. Two types of reference curves were considered: unconditional curves were dependent on age alone, whereas conditional curves were dependent on age and other covariates, namely sex and education.
Results
Conditioning on education was necessary for the Symbol Digit, Stroop Word, Stroop Color, Stroop Interference, and composite UHDRS. Unconditional curves were sufficient for the Total Motor Score. TFC was unique in that the curve was constant over age with its intercept at the maximum score (TFC = 13). For all measures, sex effects were minimal, so conditioning on sex was unwarranted.
Conclusions
Extreme quantile estimates for each measure can be considered as boundaries for natural aging and scores falling beyond these thresholds are likely the result of disease progression. Normative curves and tables are developed and can serve as references for clinical characterization in HD.
Collapse
Affiliation(s)
- James A Mills
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Jeffrey D Long
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| | | | | | | |
Collapse
|
83
|
Sewell DK, Penney J, Jay M, Zhang Y, Paulsen JS. Predicting an optimal composite outcome variable for Huntington's disease clinical trials. J Appl Stat 2020; 48:1339-1348. [PMID: 34024983 DOI: 10.1080/02664763.2020.1759034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
While there is no known cure for Huntington's disease (HD), there are early-phase clinical trials aimed at altering disease progression patterns. There is, however, no obvious single outcome for these trials to evaluate treatment efficacy. Currently used outcomes are, while reasonable, not optimal in any sense. In this paper we derive a method for constructing a composite variable via a linear combination of clinical measures. Our composite variable optimizes the signal-to-noise ratio (SNR) within the context of a longitudinal study design. We also demonstrate how to induce sparsity using a soft-approximation of an L 1 penalty on the coefficients of the composite variable. We applied our method to data from the TRACK-HD study, a longitudinal study aimed at establishing good outcome measures for HD, and found that compared to the existing composite measurement our composite variable provides a larger SNR and allows clinical trials with smaller sample sizes to achieve equivalent power.
Collapse
Affiliation(s)
- Daniel K Sewell
- Department of Biostatistics, University of Iowa, Iowa City, IA, 52242, USA
| | - Journey Penney
- Department of Biostatistics, University of Iowa, Iowa City, IA, 52242, USA
| | - Melissa Jay
- Department of Biostatistics, University of Iowa, Iowa City, IA, 52242, USA
| | - Ying Zhang
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jane S Paulsen
- Department of Psychiatry, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
84
|
Horton MC, Nopoulos P, Nance M, Landwehrmyer GB, Barker RA, Squitieri F, Burgunder JM, Quarrell O. Assessment of the Performance of a Modified Motor Scale as Applied to Juvenile Onset Huntington's Disease. J Huntingtons Dis 2020; 8:181-193. [PMID: 30856116 DOI: 10.3233/jhd-180306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Huntington's disease can present at almost any age but traditionally, those with an onset ≤20 years are described as having juvenile onset Huntington's disease (JOHD). They are more likely to have bradykinesia and dystonia earlier in the course of the disease. The Total Motor Score of the Unified Huntington's Disease Rating Scale (UHDRS-TMS) is often used as the principal outcome measure in clinical trials. OBJECTIVE To identify a motor scale more suitable for JOHD patients. METHODS A working group reviewed the UHDRS-TMS and modified it by adding four further assessment items. Rasch analysis was used to study the performance of the modified scale in 95 patients with a mean age of 19.4 (SD 6.6) years. RESULTS The initial analysis showed a significant overall misfit to the Rasch model and a number of individual items displayed poor measurement properties: all items relating to chorea displayed significant misfit due to under-discrimination. Additionally, a number of items displayed disordered response category thresholds, and a large amount of dependency was present within the item set (96 out of 741 pairwise differences = 13%). An iterative process of scale re-structuring and evaluation was then undertaken, with a view to eliminating the largest sources of misfit and generating a set of items that would conform to Rasch model expectations. CONCLUSION This post-hoc scale restructuring appears to provide a valid motor score that is psychometrically robust in a JOHD population. This scale restructuring offers a pragmatic solution to measuring motor function in a JOHD population, and it could provide the basis for the further iterative development of a more useful clinical rating scale for patients with JOHD.
Collapse
Affiliation(s)
- Mike C Horton
- Academic Department of Rehabilitation Medicine, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Peggy Nopoulos
- Departments of Psychiatry, Pediatrics and Neurology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Martha Nance
- Struthers Parkinson's Center, Minneapolis, MN, USA
| | | | - Roger A Barker
- Department of Clinical Neuroscience, John Van Geest Centre for Brain Repair, Forvie Site, Robinson Way, Cambridge, UK
| | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, IRCCS Casa Sollievo della Sofferenza Hospital (Rome-CSS Mendel), San Giovanni Rotondo, Italy
| | | | - Jean-Marc Burgunder
- Neurozentrum Siloah and Department of Neurology, Swiss HD Center, University of Bern, Switzerland
| | - Oliver Quarrell
- Department of Clinical Genetics, Sheffield Children's Hospital, Sheffield, UK
| |
Collapse
|
85
|
Marxreiter F, Stemick J, Kohl Z. Huntingtin Lowering Strategies. Int J Mol Sci 2020; 21:ijms21062146. [PMID: 32245050 PMCID: PMC7139361 DOI: 10.3390/ijms21062146] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 12/22/2022] Open
Abstract
Trials using antisense oligonucleotide technology to lower Huntingtin levels in Huntington’s disease (HD) are currently ongoing. This progress, taking place only 27 years after the identification of the Huntingtin gene (HTT) in 1993 reflects the enormous development in genetic engineering in the last decades. It is also the result of passionate basic scientific work and large worldwide registry studies that have advanced the understanding of HD. Increased knowledge of the pathophysiology of this autosomal dominantly inherited CAG-repeat expansion mediated neurodegenerative disease has led to the development of several putative treatment strategies, currently under investigation. These strategies span the whole spectrum of potential targets from genome editing via RNA interference to promoting protein degradation. Yet, recent studies revealed the importance of huntingtin RNA in the pathogenesis of the disease. Therefore, huntingtin-lowering by means of RNA interference appears to be a particular promising strategy. As a matter of fact, these approaches have entered, or are on the verge of entering, the clinical trial period. Here, we provide an overview of huntingtin-lowering approaches via DNA or RNA interference in present clinical trials as well as strategies subject to upcoming therapeutic options. We furthermore discuss putative implications for future treatment of HD patients.
Collapse
Affiliation(s)
- Franz Marxreiter
- Huntington’s Disease Outpatient Clinic, Department of Molecular Neurology, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany;
- Center for Rare Movement Disorders, Department of Molecular Neurology, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany
- Correspondence:
| | - Judith Stemick
- Huntington’s Disease Outpatient Clinic, Department of Molecular Neurology, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany;
| | - Zacharias Kohl
- Department of Neurology, University of Regensburg, Universitätsstraße 84, 93053 Regensburg, Germany;
| |
Collapse
|
86
|
Glidden AM, Luebbe EA, Elson MJ, Goldenthal SB, Snyder CW, Zizzi CE, Dorsey ER, Heatwole CR. Patient-reported impact of symptoms in Huntington disease: PRISM-HD. Neurology 2020; 94:e2045-e2053. [PMID: 32193209 DOI: 10.1212/wnl.0000000000008906] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 11/20/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine the frequency and relative importance of symptoms experienced by adults with Huntington disease (HD) and to identify factors associated with a higher disease burden. METHODS We performed 40 qualitative interviews (n = 20 with HD, n = 20 caregivers) and analyzed 2,082 quotes regarding the symptomatic burden of HD. We subsequently performed a cross-sectional study with 389 participants (n = 156 with HD [60 of whom were prodromal], n = 233 caregivers) to assess the prevalence and relative importance (scale 0-4) of 216 symptoms and 15 symptomatic themes in HD. Cross-correlation analysis was performed based on sex, disease duration, age, number of CAG repeats, disease burden, Total Functional Capacity score, employment status, disease status, and ambulatory status. RESULTS The symptomatic themes with the highest prevalence in HD were emotional issues (83.0%), fatigue (82.5%), and difficulty thinking (77.0%). The symptomatic themes with the highest relative importance to participants were difficulty thinking (1.91), impaired sleep or daytime sleepiness (1.90), and emotional issues (1.81). High Total Functional Capacity scores, being employed, and having prodromal HD were associated with a lower prevalence of symptomatic themes. Despite reporting no clinical features of the disease, prodromal individuals demonstrated high rates of emotional issues (71.2%) and fatigue (69.5%). There was concordance between the prevalence of symptoms reported by manifest individuals and caregivers. CONCLUSIONS Many symptomatic themes affect the lives of those with HD. These themes have a variable level of importance to the HD population and are identified both by those with HD and by their caregivers.
Collapse
Affiliation(s)
- Alistair M Glidden
- From the Center for Health + Technology (A.M.G., C.W.S., C.E.Z., E.R.D., C.R.H.) and Department of Neurology (E.A.L., E.R.D., C.R.H.), University of Rochester Medical Center, NY; Emory School of Medicine (M.J.E.), Emory University, Atlanta, GA; and University of Michigan Medical School (S.B.G.), University of Michigan, Ann Arbor
| | - Elizabeth A Luebbe
- From the Center for Health + Technology (A.M.G., C.W.S., C.E.Z., E.R.D., C.R.H.) and Department of Neurology (E.A.L., E.R.D., C.R.H.), University of Rochester Medical Center, NY; Emory School of Medicine (M.J.E.), Emory University, Atlanta, GA; and University of Michigan Medical School (S.B.G.), University of Michigan, Ann Arbor
| | - Molly J Elson
- From the Center for Health + Technology (A.M.G., C.W.S., C.E.Z., E.R.D., C.R.H.) and Department of Neurology (E.A.L., E.R.D., C.R.H.), University of Rochester Medical Center, NY; Emory School of Medicine (M.J.E.), Emory University, Atlanta, GA; and University of Michigan Medical School (S.B.G.), University of Michigan, Ann Arbor
| | - Steven B Goldenthal
- From the Center for Health + Technology (A.M.G., C.W.S., C.E.Z., E.R.D., C.R.H.) and Department of Neurology (E.A.L., E.R.D., C.R.H.), University of Rochester Medical Center, NY; Emory School of Medicine (M.J.E.), Emory University, Atlanta, GA; and University of Michigan Medical School (S.B.G.), University of Michigan, Ann Arbor
| | - Christopher W Snyder
- From the Center for Health + Technology (A.M.G., C.W.S., C.E.Z., E.R.D., C.R.H.) and Department of Neurology (E.A.L., E.R.D., C.R.H.), University of Rochester Medical Center, NY; Emory School of Medicine (M.J.E.), Emory University, Atlanta, GA; and University of Michigan Medical School (S.B.G.), University of Michigan, Ann Arbor
| | - Christine E Zizzi
- From the Center for Health + Technology (A.M.G., C.W.S., C.E.Z., E.R.D., C.R.H.) and Department of Neurology (E.A.L., E.R.D., C.R.H.), University of Rochester Medical Center, NY; Emory School of Medicine (M.J.E.), Emory University, Atlanta, GA; and University of Michigan Medical School (S.B.G.), University of Michigan, Ann Arbor
| | - E Ray Dorsey
- From the Center for Health + Technology (A.M.G., C.W.S., C.E.Z., E.R.D., C.R.H.) and Department of Neurology (E.A.L., E.R.D., C.R.H.), University of Rochester Medical Center, NY; Emory School of Medicine (M.J.E.), Emory University, Atlanta, GA; and University of Michigan Medical School (S.B.G.), University of Michigan, Ann Arbor
| | - Chad R Heatwole
- From the Center for Health + Technology (A.M.G., C.W.S., C.E.Z., E.R.D., C.R.H.) and Department of Neurology (E.A.L., E.R.D., C.R.H.), University of Rochester Medical Center, NY; Emory School of Medicine (M.J.E.), Emory University, Atlanta, GA; and University of Michigan Medical School (S.B.G.), University of Michigan, Ann Arbor.
| |
Collapse
|
87
|
Wijeratne PA, Johnson EB, Eshaghi A, Aksman L, Gregory S, Johnson HJ, Poudel GR, Mohan A, Sampaio C, Georgiou-Karistianis N, Paulsen JS, Tabrizi SJ, Scahill RI, Alexander DC. Robust Markers and Sample Sizes for Multicenter Trials of Huntington Disease. Ann Neurol 2020; 87:751-762. [PMID: 32105364 PMCID: PMC7187160 DOI: 10.1002/ana.25709] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/20/2023]
Abstract
Objective The identification of sensitive biomarkers is essential to validate therapeutics for Huntington disease (HD). We directly compare structural imaging markers across the largest collective imaging HD dataset to identify a set of imaging markers robust to multicenter variation and to derive upper estimates on sample sizes for clinical trials in HD. Methods We used 1 postprocessing pipeline to retrospectively analyze T1‐weighted magnetic resonance imaging (MRI) scans from 624 participants at 3 time points, from the PREDICT‐HD, TRACK‐HD, and IMAGE‐HD studies. We used mixed effects models to adjust regional brain volumes for covariates, calculate effect sizes, and simulate possible treatment effects in disease‐affected anatomical regions. We used our model to estimate the statistical power of possible treatment effects for anatomical regions and clinical markers. Results We identified a set of common anatomical regions that have similarly large standardized effect sizes (>0.5) between healthy control and premanifest HD (PreHD) groups. These included subcortical, white matter, and cortical regions and nonventricular cerebrospinal fluid (CSF). We also observed a consistent spatial distribution of effect size by region across the whole brain. We found that multicenter studies were necessary to capture treatment effect variance; for a 20% treatment effect, power of >80% was achieved for the caudate (n = 661), pallidum (n = 687), and nonventricular CSF (n = 939), and, crucially, these imaging markers provided greater power than standard clinical markers. Interpretation Our findings provide the first cross‐study validation of structural imaging markers in HD, supporting the use of these measurements as endpoints for both observational studies and clinical trials. ANN NEUROL 2020;87:751–762
Collapse
Affiliation(s)
- Peter A Wijeratne
- Center for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Eileanoir B Johnson
- Huntington's Disease Research Center, Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, United Kingdom
| | - Arman Eshaghi
- Center for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom.,Queen Square Multiple Sclerosis Center, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Leon Aksman
- Center for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| | - Sarah Gregory
- Huntington's Disease Research Center, Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, United Kingdom
| | - Hans J Johnson
- Departments of Neurology and Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA.,Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA
| | - Govinda R Poudel
- Mary Mackillop Institute of Health Research, Australian Catholic University, Melbourne, Australia
| | | | | | - Nellie Georgiou-Karistianis
- Monash Institute of Cognitive and Clinical Neurosciences, School of Psychological Sciences, Faculty of Nursing, Medicine, and Health Sciences, Monash University, Clayton Campus, Victoria, Australia
| | - Jane S Paulsen
- Departments of Neurology and Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Sarah J Tabrizi
- Huntington's Disease Research Center, Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, United Kingdom
| | - Rachael I Scahill
- Huntington's Disease Research Center, Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, United Kingdom
| | | | - Daniel C Alexander
- Center for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom
| |
Collapse
|
88
|
Rodrigues FB, Quinn L, Wild EJ. Huntington's Disease Clinical Trials Corner: January 2019. J Huntingtons Dis 2020; 8:115-125. [PMID: 30776019 DOI: 10.3233/jhd-190001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this edition of the Huntington's Disease Clinical Trials Corner we expand on the GENERATION-HD1 and PACE-HD trials, and we list all currently registered and ongoing clinical trials in Huntington's disease.
Collapse
Affiliation(s)
- Filipe B Rodrigues
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, UK.,Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Medicine, University of Lisbon, PT.,Clinical Pharmacology Unit, Instituto de Medicina Molecular, Lisbon, PT
| | - Lori Quinn
- Department of Biobehavioral Sciences, Teachers College, Columbia University, USA
| | - Edward J Wild
- UCL Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, UK
| |
Collapse
|
89
|
McGarry A, Leinonen M, Kieburtz K, Geva M, Olanow CW, Hayden M. Effects of Pridopidine on Functional Capacity in Early-Stage Participants from the PRIDE-HD Study. J Huntingtons Dis 2020; 9:371-380. [PMID: 33164941 PMCID: PMC7836066 DOI: 10.3233/jhd-200440] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND No pharmacological treatment has been demonstrated to provide a functional benefit for persons with Huntington's disease (HD). Pridopidine is a sigma-1-receptor agonist shown to have beneficial effects in preclinical models of HD. OBJECTIVE To further explore the effect of pridopidine on Total Functional Capacity (TFC) in the recent double-blind, placebo-controlled PRIDE-HD study. METHODS We performed post-hoc analyses to evaluate the effect of pridopidine on TFC at 26 and 52 weeks. Participants were stratified according to baseline TFC score and analyzed using repeated measures (MMRM) and multiple imputation assuming missing not-at-random (MNAR) and worst-case scenarios. RESULTS The pridopidine 45 mg bid dosage demonstrated a beneficial effect on TFC for the entire population at week 52 of 0.87 (nominal p = 0.0032). The effect was more pronounced for early HD participants (HD1/HD2, TFC = 7-13), with a change from placebo of 1.16 (nominal p = 0.0003). This effect remained nominally significant using multiple imputation with missing not at random assumption as a sensitivity analysis. Responder analyses showed pridopidine 45 mg bid reduced the probability of TFC decline in early HD patients at Week 52 (nominal p = 0.02). CONCLUSION Pridopidine 45 mg bid results in a nominally significant reduction in TFC decline at 52 weeks compared to placebo, particularly in patients with early-stage HD.
Collapse
Affiliation(s)
- Andrew McGarry
- Cooper University Healthcare at Rowan University, Camden, NJ, USA
- Clintrex Research Corporation, Sarasota, FL, USA
| | | | | | | | - C. Warren Olanow
- Mount Sinai School of Medicine, New York City, NY, USA
- Clintrex Research Corporation, Sarasota, FL, USA
| | - Michael Hayden
- Prilenia Therapeutics, Herzliya, Israel
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
90
|
Zhao Y, Yu Q, Lake SL. A flexible multi-domain test with adaptive weights and its application to clinical trials. Pharm Stat 2019; 19:315-325. [PMID: 31886602 DOI: 10.1002/pst.1993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/30/2019] [Accepted: 11/21/2019] [Indexed: 12/20/2022]
Abstract
The design of a clinical trial is often complicated by the multi-systemic nature of the disease; a single endpoint often cannot capture the spectrum of potential therapeutic benefits. Multi-domain outcomes which take into account patient heterogeneity of disease presentation through measurements of multiple symptom/functional domains are an attractive alternative to a single endpoint. A multi-domain test with adaptive weights is proposed to synthesize the evidence of treatment efficacy over numerous disease domains. The test is a weighted sum of domain-specific test statistics with weights selected adaptively via a data-driven algorithm. The null distribution of the test statistic is constructed empirically through resampling and does not require estimation of the covariance structure of domain-specific test statistics. Simulations show that the proposed test controls the type I error rate, and has increased power over other methods such as the O'Brien and Wei-Lachin tests in scenarios reflective of clinical trial settings. Data from a clinical trial in a rare lysosomal storage disorder were used to illustrate the properties of the proposed test. As a strategy of combining marginal test statistics, the proposed test is flexible and readily applicable to a variety of clinical trial scenarios.
Collapse
Affiliation(s)
- Yang Zhao
- Gilead Sciences, Foster City, California
| | - Qifeng Yu
- Sanofi R&D, Framingham, Massachusetts
| | | |
Collapse
|
91
|
Abnormal Photic Entrainment to Phase-Delaying Stimuli in the R6/2 Mouse Model of Huntington's Disease, despite Retinal Responsiveness to Light. eNeuro 2019; 6:ENEURO.0088-19.2019. [PMID: 31744839 PMCID: PMC6905640 DOI: 10.1523/eneuro.0088-19.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 10/22/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022] Open
Abstract
The circadian clock located in the suprachiasmatic nucleus (SCN) in mammals entrains to ambient light via the retinal photoreceptors. This allows behavioral rhythms to change in synchrony with seasonal and daily changes in light period. Circadian rhythmicity is progressively disrupted in Huntington's disease (HD) and in HD mouse models such as the transgenic R6/2 line. Although retinal afferent inputs to the SCN are disrupted in R6/2 mice at late stages, they can respond to changes in light/dark cycles, as seen in jet lag and 23 h/d paradigms. To investigate photic entrainment and SCN function in R6/2 mice at different stages of disease, we first assessed the effect on locomotor activity of exposure to a 15 min light pulse given at different times of the day. We then placed the mice under five non-standard light conditions. These were light cycle regimes (T-cycles) of T21 (10.5 h light/dark), T22 (11 h light/dark), T26 (13 h light/dark), constant light, or constant dark. We found a progressive impairment in photic synchronization in R6/2 mice when the stimuli required the SCN to lengthen rhythms (phase-delaying light pulse, T26, or constant light), but normal synchronization to stimuli that required the SCN to shorten rhythms (phase-advancing light pulse and T22). Despite the behavioral abnormalities, we found that Per1 and c-fos gene expression remained photo-inducible in SCN of R6/2 mice. Both the endogenous drift of the R6/2 mouse SCN to shorter periods and its inability to adapt to phase-delaying changes will contribute to the HD circadian dysfunction.
Collapse
|
92
|
Langbehn DR, Stout JC, Gregory S, Mills JA, Durr A, Leavitt BR, Roos RAC, Long JD, Owen G, Johnson HJ, Borowsky B, Craufurd D, Reilmann R, Landwehrmeyer GB, Scahill RI, Tabrizi SJ. Association of CAG Repeats With Long-term Progression in Huntington Disease. JAMA Neurol 2019; 76:1375-1385. [PMID: 31403680 PMCID: PMC6692683 DOI: 10.1001/jamaneurol.2019.2368] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/02/2019] [Indexed: 11/14/2022]
Abstract
IMPORTANCE In Huntington disease (HD), mutation severity is defined by the length of the CAG trinucleotide sequence, a well-known predictor of clinical onset age. The association with disease trajectory is less well characterized. Quantifiable summary measures of trajectory applicable over decades of early disease progression are lacking. An accurate model of the age-CAG association with early progression is critical to clinical trial design, informing both sample size and intervention timing. OBJECTIVE To succinctly capture the decades-long early progression of HD and its dependence on CAG repeat length. DESIGN, SETTING, AND PARTICIPANTS Prospective study at 4 academic HD treatment and research centers. Participants were the combined sample from the TRACK-HD and Track-On HD studies consisting of 290 gene carriers (presymptomatic to stage II), recruited from research registries at participating centers, and 153 nonbiologically related controls, generally spouses or friends. Recruitment was targeted to match a balanced, prespecified spectrum of age, CAG repeat length, and diagnostic status. In the TRACK-HD and Track-On HD studies, 13 and 5 potential participants, respectively, failed study screening. Follow-up ranged from 0 to 6 years. The study dates were January 2008 to November 2014. These analyses were performed between December 2015 and January 2019. MAIN OUTCOMES AND MEASURES The outcome measures were principal component summary scores of motor-cognitive function and of brain volumes. The main outcome was the association of these scores with age and CAG repeat length. RESULTS We analyzed 2065 visits from 443 participants (247 female [55.8%]; mean [SD] age, 44.4 [10.3] years). Motor-cognitive measures were highly correlated and had similar CAG repeat length-dependent associations with age. A composite summary score accounted for 67.6% of their combined variance. This score was well approximated by a score combining 3 items (total motor score, Symbol Digit Modalities Test, and Stroop word reading) from the Unified Huntington's Disease Rating Scale. For either score, initial progression age and then acceleration rate were highly CAG repeat length dependent. The acceleration continues through at least stage II disease. In contrast, 3 distinct patterns emerged among brain measures (basal ganglia, gray matter, and a combination of whole-brain, ventricular, and white matter volumes). The basal ganglia pattern showed considerable change in even the youngest participants but demonstrated minimal acceleration of loss with aging. Each clinical and brain summary score was strongly associated with the onset and rate of decline in total functional capacity. CONCLUSIONS AND RELEVANCE Results of this study suggest that succinct summary measures of function and brain loss characterize HD progression across a wide disease span. CAG repeat length strongly predicts their decline rate. This work aids our understanding of the age and CAG repeat length-dependent association between changes in the brain and clinical manifestations of HD.
Collapse
Affiliation(s)
| | - Julie C. Stout
- School of Psychology and Psychiatry, Monash University, Melbourne, Victoria, Australia
| | - Sarah Gregory
- Huntington’s Disease Centre, UCL Institute of Neurology, University College London, Queen Square, London, United Kingdom
| | | | - Alexandra Durr
- Institut du Cerveau et de la Moelle Epinière (ICM), Genetic Department, Assistance Publique–Hôpitaux de Paris, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale Unité 1127, Le Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 7225, Pitié-Salpêtrière University Hospital, Paris, France
| | - Blair R. Leavitt
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Raymund A. C. Roos
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Gail Owen
- Huntington’s Disease Centre, UCL Institute of Neurology, University College London, Queen Square, London, United Kingdom
| | - Hans J. Johnson
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City
| | | | - David Craufurd
- Manchester Academic Health Sciences Centre, Central Manchester University Hospitals National Health Service Foundation Trust, University of Manchester, Manchester, United Kingdom
| | - Ralf Reilmann
- George-Huntington-Institute, Department of Radiology, University of Münster, Münster, Germany
- Hertie-Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | | | - Rachael I. Scahill
- Huntington’s Disease Centre, UCL Institute of Neurology, University College London, Queen Square, London, United Kingdom
| | - Sarah J. Tabrizi
- Huntington’s Disease Centre, UCL Institute of Neurology, University College London, Queen Square, London, United Kingdom
| |
Collapse
|
93
|
Garcia TP, Wang Y, Shoulson I, Paulsen JS, Marder K. Disease Progression in Huntington Disease: An Analysis of Multiple Longitudinal Outcomes. J Huntingtons Dis 2019; 7:337-344. [PMID: 30400103 DOI: 10.3233/jhd-180297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Critical to discovering targeted therapies for Huntington disease (HD) are validated methods that more precisely predict when clinical outcomes occur for different patient profiles. OBJECTIVE To more precisely predict the probability of when motor diagnosis (diagnostic confidence level 4) on the Unified Huntington's Disease Rating Scale (UHDRS), cognitive impairment (two or more neuropsychological scores on the UHDRS were 1.5 standard deviations below normative means) and Stage II Total Functional Capacity (TFC) first occur by accounting for dependencies between these outcomes. METHODS Adult premanifest participants with ≥36 CAG repeats were selected from multi-center, longitudinal, observational studies: Prospective Huntington At Risk Observational Study (PHAROS, n = 346), Neurobiological Predictors of Huntington Disease (PREDICT, n = 909); and Cooperative Huntington Observational Research Trial (COHORT, n = 430). Probabilities were estimated for each study, and pooled using the Joint Progression of Risk Assessment Tool (JPRAT) which accounts for dependencies between outcomes. RESULTS All studies had similar probabilities of when motor diagnosis, cognitive impairment, and Stage II TFC first occurred. Probability estimates from JPRAT were 43% less variable than from models that ignored dependencies between outcomes. The probability of experiencing motor-diagnosis, cognitive impairment, and Stage II TFC within 5 years was 10%, 18%, and 7%, respectively for 45-year-olds with 42 CAG repeats, and was 4%, 10% and 5%, respectively, for 40 year olds with 42 CAG repeats. CONCLUSIONS Improved predictions from JPRAT may benefit treatment studies of rare diseases and is an alternative to composite outcomes when the objective is interpreting individual outcomes within the same model.
Collapse
Affiliation(s)
- Tanya P Garcia
- Department of Statistics, Texas A&M University, College Station, TX, USA
| | - Yuanjia Wang
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Ira Shoulson
- Department of Neurology, Georgetown University, Washington, DC, USA
| | - Jane S Paulsen
- Department of Psychological and Brain Sciences, The University of Iowa, Iowa City, IA, USA
| | - Karen Marder
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
94
|
Drew CJG, Quinn L, Hamana K, Williams-Thomas R, Marsh L, Dimitropoulou P, Playle R, Griffin BA, Kelson M, Schubert R, Muratori L, Reilmann R, Rosser A, Busse M. Physical Activity and Exercise Outcomes in Huntington Disease (PACE-HD): Protocol for a 12-Month Trial Within Cohort Evaluation of a Physical Activity Intervention in People With Huntington Disease. Phys Ther 2019; 99:1201-1210. [PMID: 31101920 DOI: 10.1093/ptj/pzz075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 02/18/2019] [Indexed: 02/09/2023]
Abstract
BACKGROUND Exercise is emerging as an important aspect in the management of disease-related symptoms and functional decline in people with Huntington disease (HD). Long-term evaluation of physical activity and exercise participation in HD has yet to be undertaken. OBJECTIVE The objective is to investigate the feasibility of a nested randomized controlled trial (RCT) alongside a longitudinal observational study of physical activity and exercise outcomes in people with HD. DESIGN This will be a 12-month longitudinal observational study (n = 120) with a nested evaluation of a physical activity intervention (n = 30) compared with usual activity (n = 30) using a "trial within a cohort" design. SETTING The study will take place in HD specialist clinics in Germany, Spain, and the United States, with intervention delivery in community settings. PARTICIPANTS The participants will have early-mid-stage HD and be participating in the Enroll-HD study. INTERVENTION This will be a 12-month physical activity behavioral change intervention, delivered by physical therapists in 18 sessions, targeting uptake of aerobic exercise and increased physical activity. MEASUREMENTS All participants (n = 120) will complete Enroll-HD assessments (motor, cognitive, behavioral, and quality of life) at baseline and at 12 months. Additional Physical ACtivity and Exercise Outcomes in Huntington Disease (PACE-HD) assessments include fitness (predicted maximal oxygen uptake [V o2max]), self-reported and quantitative measures of physical activity, disease-specific symptoms, and walking endurance. RCT participants (n = 60) will complete an additional battery of quantitative motor assessments and a 6-month interim assessment. Enroll-HD data will be linked to PACE-HD physical activity and fitness data. LIMITATIONS The limitations include that the embedded RCT is open, and assessors at RCT sites are not blinded to participant allocation. CONCLUSION PACE-HD will enable determination of the feasibility of long-term physical activity interventions in people with HD. The novel "trial within a cohort" design and incorporation of data linkage have potential to reduce participant burden. This design could be applied to other neurological diseases and movement disorders where recruitment and retention are challenging.
Collapse
Affiliation(s)
- Cheney J G Drew
- Centre for Trials Research, Cardiff University, South Glamorgan, United Kingdom
| | - Lori Quinn
- Centre for Trials Research, Cardiff University; and Teachers College, Columbia University, New York, New York
| | - Katy Hamana
- School of Health Care Sciences, Cardiff University
| | | | - Lucy Marsh
- Centre for Trials Research, Cardiff University, South Glamorgan, United Kingdom
| | | | - Rebecca Playle
- Centre for Trials Research, Cardiff University, South Glamorgan, United Kingdom
| | | | - Mark Kelson
- Department of Mathematics, University of Exeter, Exeter, United Kingdom
| | - Robin Schubert
- George Huntington Institute and Institute for Clinical Radiology, University of Münster, Münster, Germany
| | - Lisa Muratori
- George Huntington Institute and Institute for Clinical Radiology, University of Münster; and School of Health Technology and Management, Stony Brook University, Stony Brook, New York
| | - Ralf Reilmann
- George Huntington Institute and Institute for Clinical Radiology, University of Münster; and Department of Neurodegenerative Diseases and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | | | - Monica Busse
- Centre for Trials Research, Cardiff University, 4th Floor, Neuadd Meirionydd Health Park, Cardiff, South Glamorgan CF14 7YS, United Kingdom
| |
Collapse
|
95
|
Playle R, Dimitropoulou P, Kelson M, Quinn L, Busse M. Exercise Interventions in Huntington's Disease: An Individual Patient Data Meta-Analysis. Mov Disord Clin Pract 2019; 6:567-575. [PMID: 31538091 DOI: 10.1002/mdc3.12809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 05/03/2019] [Accepted: 06/07/2019] [Indexed: 12/18/2022] Open
Abstract
Background Physical activity may be beneficial in Huntington's disease (HD); however, studies to date have been underpowered to detect change. We combined data from five randomized controlled feasibility trials using individual patient data meta-analyses. Methods/Design All trial interventions comprised a combination of supervised and self-directed physical activity, with varied emphasis on aerobic, strength, endurance, flexibility, and task training. Duration ranged from 8 to 16 weeks. The primary outcome was the modified Unified Huntington's Disease Rating Motor Score. Secondary outcomes included the Symbol Digit Modality Test, Berg Balance Scale, 30-second Chair stand, Timed Up and Go, Gait Speed, Physical Performance Test, Six-Minute Walk, International Physical Activity Questionnaire, Hospital Anxiety and Depression Scale, EuroQol Health Utility Index, and Short-Form 36 Health Related Quality of Life Scale. The primary analysis utilized a two-stage approach. A one-stage approach was explored as a sensitivity analysis using a cross-classified (by study site) linear mixed-effects model. Results One hundred twenty-one participants provided complete data. Risk of bias was moderate; however, primary outcomes were blind assessed. Primary pooled effect estimates adjusted for baseline modified motor score (95% confidence interval) were 0.2 (-2.1 to 2.6) favoring control. There was considerable heterogeneity between the studies. Conclusions There was no evidence of an exercise effect on the modified motor score in these relatively short-duration interventions. Longer-duration trials incorporating supervised components meeting frequency, intensity, time, and type principles are required. Lack of common outcomes limited the analysis and highlight the importance of a core outcome set for evaluating exercise in HD.
Collapse
Affiliation(s)
- Rebecca Playle
- Centre for Trials Research Cardiff University Cardiff United Kingdom
| | | | - Mark Kelson
- School of Mathematics/The Alan Turing Institute University of Exeter Exeter United Kingdom
| | - Lori Quinn
- Centre for Trials Research Cardiff University Cardiff United Kingdom.,Teachers College Columbia University New York New York USA
| | - Monica Busse
- Centre for Trials Research Cardiff University Cardiff United Kingdom
| |
Collapse
|
96
|
Harrer S, Shah P, Antony B, Hu J. Artificial Intelligence for Clinical Trial Design. Trends Pharmacol Sci 2019; 40:577-591. [PMID: 31326235 DOI: 10.1016/j.tips.2019.05.005] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 05/28/2019] [Accepted: 05/28/2019] [Indexed: 12/23/2022]
Abstract
Clinical trials consume the latter half of the 10 to 15 year, 1.5-2.0 billion USD, development cycle for bringing a single new drug to market. Hence, a failed trial sinks not only the investment into the trial itself but also the preclinical development costs, rendering the loss per failed clinical trial at 800 million to 1.4 billion USD. Suboptimal patient cohort selection and recruiting techniques, paired with the inability to monitor patients effectively during trials, are two of the main causes for high trial failure rates: only one of 10 compounds entering a clinical trial reaches the market. We explain how recent advances in artificial intelligence (AI) can be used to reshape key steps of clinical trial design towards increasing trial success rates.
Collapse
Affiliation(s)
- Stefan Harrer
- IBM Research, IBM Research Australia Lab, 3006 Melbourne, VIC, Australia.
| | - Pratik Shah
- Massachusetts Institute of Technology, Media Lab, 02139 Cambridge, MA, USA
| | - Bhavna Antony
- IBM Research, IBM Research Australia Lab, 3006 Melbourne, VIC, Australia
| | - Jianying Hu
- IBM Research, IBM T.J. Watson Research Center, 10598 Yorktown Heights, NY, USA
| |
Collapse
|
97
|
Tabrizi SJ, Leavitt BR, Landwehrmeyer GB, Wild EJ, Saft C, Barker RA, Blair NF, Craufurd D, Priller J, Rickards H, Rosser A, Kordasiewicz HB, Czech C, Swayze EE, Norris DA, Baumann T, Gerlach I, Schobel SA, Paz E, Smith AV, Bennett CF, Lane RM. Targeting Huntingtin Expression in Patients with Huntington's Disease. N Engl J Med 2019; 380:2307-2316. [PMID: 31059641 DOI: 10.1056/nejmoa1900907] [Citation(s) in RCA: 463] [Impact Index Per Article: 77.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Huntington's disease is an autosomal-dominant neurodegenerative disease caused by CAG trinucleotide repeat expansion in HTT, resulting in a mutant huntingtin protein. IONIS-HTTRx (hereafter, HTTRx) is an antisense oligonucleotide designed to inhibit HTT messenger RNA and thereby reduce concentrations of mutant huntingtin. METHODS We conducted a randomized, double-blind, multiple-ascending-dose, phase 1-2a trial involving adults with early Huntington's disease. Patients were randomly assigned in a 3:1 ratio to receive HTTRx or placebo as a bolus intrathecal administration every 4 weeks for four doses. Dose selection was guided by a preclinical model in mice and nonhuman primates that related dose level to reduction in the concentration of huntingtin. The primary end point was safety. The secondary end point was HTTRx pharmacokinetics in cerebrospinal fluid (CSF). Prespecified exploratory end points included the concentration of mutant huntingtin in CSF. RESULTS Of the 46 patients who were enrolled in the trial, 34 were randomly assigned to receive HTTRx (at ascending dose levels of 10 to 120 mg) and 12 were randomly assigned to receive placebo. Each patient received all four doses and completed the trial. Adverse events, all of grade 1 or 2, were reported in 98% of the patients. No serious adverse events were seen in HTTRx-treated patients. There were no clinically relevant adverse changes in laboratory variables. Predose (trough) concentrations of HTTRx in CSF showed dose dependence up to doses of 60 mg. HTTRx treatment resulted in a dose-dependent reduction in the concentration of mutant huntingtin in CSF (mean percentage change from baseline, 10% in the placebo group and -20%, -25%, -28%, -42%, and -38% in the HTTRx 10-mg, 30-mg, 60-mg, 90-mg, and 120-mg dose groups, respectively). CONCLUSIONS Intrathecal administration of HTTRx to patients with early Huntington's disease was not accompanied by serious adverse events. We observed dose-dependent reductions in concentrations of mutant huntingtin. (Funded by Ionis Pharmaceuticals and F. Hoffmann-La Roche; ClinicalTrials.gov number, NCT02519036.).
Collapse
Affiliation(s)
- Sarah J Tabrizi
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Blair R Leavitt
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - G Bernhard Landwehrmeyer
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Edward J Wild
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Carsten Saft
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Roger A Barker
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Nick F Blair
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - David Craufurd
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Josef Priller
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Hugh Rickards
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Anne Rosser
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Holly B Kordasiewicz
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Christian Czech
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Eric E Swayze
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Daniel A Norris
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Tiffany Baumann
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Irene Gerlach
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Scott A Schobel
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Erika Paz
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Anne V Smith
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - C Frank Bennett
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| | - Roger M Lane
- From University College London (UCL) Huntington's Disease Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, and the U.K. Dementia Research Institute at UCL, London (S.J.T., E.J.W.), the Department of Clinical Neuroscience, Addenbrooke's Hospital, University of Cambridge, Cambridge (R.A.B., N.F.B.), Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, and the Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester (D.C.), the University of Edinburgh and the U.K. Dementia Research Institute, Edinburgh (J.P.), the Institute of Clinical Sciences, College of Medical and Dental Sciences, University Hospital Birmingham, Birmingham (H.R.), and the Cardiff University Brain Repair Group, Brain Repair and Intracranial Neurotherapeutics Unit, Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff (A.R.) - all in the United Kingdom; the Centre for Huntington's Disease, Department of Medical Genetics, and the Division of Neurology, Department of Medicine, University of British Columbia, and the Centre for Molecular Medicine and Therapeutics, B.C. Children's Hospital, Vancouver, Canada (B.R.L.); the Department of Neurology, Ulm University, Huntington's Disease Centre, Ulm (G.B.L.), the Department of Neurology, Huntington Center North Rhine-Westphalia, Ruhr University Bochum, St. Josef-Hospital, Bochum (C.S.), and the Department of Neuropsychiatry, Charité-Universitätsmedizin Berlin, Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin (J.P.) - all in Germany; Ionis Pharmaceuticals, Carlsbad, CA (H.B.K., E.E.S., D.A.N., T.B., E.P., A.V.S., C.F.B., R.M.L.); and F. Hoffmann-La Roche, Basel, Switzerland (C.C., I.G., S.A.S.)
| |
Collapse
|
98
|
Müller HP, Huppertz HJ, Dreyhaupt J, Ludolph AC, Tabrizi SJ, Roos RAC, Durr A, Landwehrmeyer GB, Kassubek J. Combined cerebral atrophy score in Huntington's disease based on atlas-based MRI volumetry: Sample size calculations for clinical trials. Parkinsonism Relat Disord 2019; 63:179-184. [PMID: 30846243 DOI: 10.1016/j.parkreldis.2019.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/18/2018] [Accepted: 02/03/2019] [Indexed: 12/19/2022]
Abstract
INTRODUCTION A volumetric MRI analysis of longitudinal regional cerebral atrophy in Huntington's disease (HD) was performed as a read-out of disease progression to calculate sample sizes for future clinical trials. METHODS This study was based on MRI data of 59 patients with HD and 40 controls recruited within the framework of the PADDINGTON study and investigated at baseline and follow-up after 6 and 15 months. Automatic atlas-based volumetry (ABV) of structural T1-weighted scans was used to calculate longitudinal volume changes of brain structures relevant in HD and to assess standardized effect sizes and sample sizes required for potential future studies. RESULTS Atrophy rates were largest in the caudate (-3.4%), putamen (-2.8%), nucleus accumbens (-1.6%), and the parietal lobes (-1.7%); the lateral ventricles showed an expansion by 6.0%. Corresponding effect sizes were -1.35 (caudate), -0.84 (putamen), -0.91 (nucleus accumbens), -1.05 (parietal lobe), and 0.92 (lateral ventricles) leading to N = 36 subjects per study group for detecting a 50% attenuation of atrophy for the best performing structure (caudate). A combined score of volume changes in non-overlapping compartments (striatum, parietal lobes, lateral ventricles) increased the effect size to -1.60 and substantially reduced the required sample sizes by 10 to N = 26 subjects per study group. This combined imaging score correlated significantly both with the CAP score and with the progression of the clinical phenotype. CONCLUSION We propose ABV of the striatum together with parietal lobe and lateral ventricle volumes as a combined imaging read-out for progression studies including clinical trials in HD.
Collapse
Affiliation(s)
| | | | - Jens Dreyhaupt
- Institute of Epidemiology and Medical Biometry, University of Ulm, Germany
| | | | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Raymund A C Roos
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Alexandra Durr
- ICM - Institut du Cerveau et de la Moelle Epinière, INSERM U1127, CNRS UMR7225, Sorbonne Universités - UPMC Université Paris VI UMR_S1127 and APHP, Genetic Department, Pitié-Salpêtrière University Hospital, Paris, France
| | | | - Jan Kassubek
- Department of Neurology, University of Ulm, Germany
| |
Collapse
|
99
|
Shahn Z, Li Y, Sun Z, Mohan A, Sampaio C, Hu J. G-Computation and Hierarchical Models for Estimating Multiple Causal Effects From Observational Disease Registries With Irregular Visits. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2019; 2019:789-798. [PMID: 31259036 PMCID: PMC6568089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Huntington's Disease (HD) is a neurodegenerative disorder with serious motor, cognitive, and behavioral symptoms. Chorea, a motor symptom of HD characterized by abrupt involuntary movements, is typically treated with tetrabenazine or certain off-label antipsychotics. Clinical trial evidence about the impact of these drugs in the HD population is scant. However, multiple observational HD registries have recently been used with success to model HD progression1,2 and provide an opportunity to obtain effect estimates in the absence of clinical trials. We use a dataset integrated from four large-scale HD registries to generate evidence on the efficacy of chorea treatments for chorea as well as their impact on other aspects of HD progression. Clinical conclusions are meant only to illustrate our methodological approach. We employ parametric G-computation for causal inference to adjust for confounding and accommodate irregular visits and treatment patterns. We fit Bayesian hierarchical models to the results of multiple related analyses to share strength across studies and handle multiple comparisons concerns.
Collapse
Affiliation(s)
- Zach Shahn
- IBM T. J. Watson Research Center, Yorktown Heights, NY, USA
| | - Ying Li
- IBM T. J. Watson Research Center, Yorktown Heights, NY, USA
| | - Zhaonan Sun
- IBM T. J. Watson Research Center, Yorktown Heights, NY, USA
| | - Amrita Mohan
- Cure Huntington's Disease Institute, Princeton, NJ, USA
| | | | - Jianying Hu
- IBM T. J. Watson Research Center, Yorktown Heights, NY, USA
| |
Collapse
|
100
|
Long JD, Mills JA. Joint modeling of multivariate longitudinal data and survival data in several observational studies of Huntington's disease. BMC Med Res Methodol 2018; 18:138. [PMID: 30445915 PMCID: PMC6240282 DOI: 10.1186/s12874-018-0592-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/29/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Joint modeling is appropriate when one wants to predict the time to an event with covariates that are measured longitudinally and are related to the event. An underlying random effects structure links the survival and longitudinal submodels and allows for individual-specific predictions. Multiple time-varying and time-invariant covariates can be included to potentially increase prediction accuracy. The goal of this study was to estimate a multivariate joint model on several longitudinal observational studies of Huntington's disease, examine external validity performance, and compute individual-specific predictions for characterizing disease progression. Emphasis was on the survival submodel for predicting the hazard of motor diagnosis. METHODS Data from four observational studies was analyzed: Enroll-HD, PREDICT-HD, REGISTRY, and Track-HD. A Bayesian approach to estimation was adopted, and external validation was performed using a time-varying AUC measure. Individual-specific cumulative hazard predictions were computed based on a simulation approach. The cumulative hazard was used for computing predicted age of motor onset and also for a deviance residual indicating the discrepancy between observed diagnosis status and model-based status. RESULTS The joint model trained in a single study had very good performance in discriminating among diagnosed and pre-diagnosed participants in the remaining test studies, with the 5-year mean AUC = .83 (range .77-.90), and the 10-year mean AUC = .86 (range .82-.92). Graphical analysis of the predicted age of motor diagnosis showed an expected strong relationship with the trinucleotide expansion that causes Huntington's disease. Graphical analysis of the deviance-type residual revealed there were individuals who converted to a diagnosis despite having relatively low model-based risk, others who had not yet converted despite having relatively high risk, and the majority falling between the two extremes. CONCLUSIONS Joint modeling is an improvement over traditional survival modeling because it considers all the longitudinal observations of covariates that are predictive of an event. Predictions from joint models can have greater accuracy because they are tailored to account for individual variability. These predictions can provide relatively accurate characterizations of individual disease progression, which might be important in the timing of interventions, determining the qualification for appropriate clinical trials, and general genotypic analysis.
Collapse
Affiliation(s)
- Jeffrey D. Long
- Department of Psychiatry, Carver College of Medicine, University of Iowa, 500 Newton Road, Iowa City, IA 52242-1000 USA
- Department of Biostatistics, Department of Public Health, University of Iowa, 145 N. Riverside Drive, Iowa City, IA 52242-1000 USA
| | - James A. Mills
- Department of Psychiatry, Carver College of Medicine, University of Iowa, 500 Newton Road, Iowa City, IA 52242-1000 USA
| |
Collapse
|