1
|
de l'Escalopier N, Voisard C, Jung S, Michaud M, Moreau A, Vayatis N, Denormandie P, Verrando A, Verdaguer C, Moussu A, Jequier A, Duret C, Mailhan L, Gatin L, Oudre L, Ricard D. Inertial measurement units to evaluate the efficacity of Equino Varus Foot surgery in post stroke hemiparetic patients: a feasibility study. J Neuroeng Rehabil 2024; 21:182. [PMID: 39407309 PMCID: PMC11481626 DOI: 10.1186/s12984-024-01469-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
INTRODUCTION This study evaluates the gait analysis obtained by Inetial Measurement Units (IMU) before and after surgical management of Spastic Equino Varus Foot (SEVF) in hemiplegic post-stroke patients and to compare it with the functional results obtained in a monocentric prospective cohort. METHODS Patients with post-stroke SEVF, who underwent surgery in a single hospital between November 2019 and December 2021 were included. The follow-up duration was 6 months and included a functional analysis using Goal Attainment Scaling (GAS) and a Gait analysis using an innovative Multidimensional Gait Evaluation using IMU: the semiogram. RESULTS 20 patients had a gait analysis preoperatively and at 6 months postoperatively. 90% (18/20) patients had a functional improvement (GAS T score ≥ 50) and 50% (10/20) had an improvement in walking technique as evidenced by the cessation of the use of a walking aid (WA). In patients with functional improvement and modification of WA the change in the semiogram area was + 9.5%, sd = 27.5%, and it was + 15.4%, sd = 28%. In the group with functional improvement without change of WA. For the 3 experiences (two patients) with unfavorable results, the area under the curve changed by + 2.3%, -10.2% and - 9.5%. The measurement of the semiogram area weighted by average speed demonstrated very good reproducibility (ICC(1, 3) = 0.80). DISCUSSION IMUs appear to be a promising solution for the assessment of post-stroke hemiplegic patients who have undergone SEVF surgery. They can provide a quantified, objective, reliable in individual longitudinal follow up automated gait analysis solution for routine clinical use. Combined with a functional scale such as the GAS, they can provide a global analysis of the effect of surgery.
Collapse
Affiliation(s)
- Nicolas de l'Escalopier
- Service de Chirurgie Orthopédique, Traumatologique et Réparatrice des Membres, Service de Santé des Armées, HIA Percy, Clamart, F-92140, France
- Université Paris Cité, Université Paris Saclay, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, Paris, France
- CRF Les Trois Soleils, Médecine Physique et de Réadaptation, Unité de Rééducation Neurologique, Boissise-Le-Roi, France
| | - Cyril Voisard
- Université Paris Cité, Université Paris Saclay, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, Paris, France
| | - Sylvain Jung
- Université Paris Saclay, Université Paris Cité, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, Gif-sur-Yvette, F-91190, France
| | - Mona Michaud
- Université Paris Cité, Université Paris Saclay, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, Paris, France
| | - Albane Moreau
- Université Paris Cité, Université Paris Saclay, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, Paris, France
| | - Nicolas Vayatis
- Université Paris Saclay, Université Paris Cité, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, Gif-sur-Yvette, F-91190, France
| | - Philippe Denormandie
- Service de Chirurgie Orthopédique, Traumatologique et Réparatrice des Membres, Service de Santé des Armées, HIA Percy, Clamart, F-92140, France
- Service de Chirurgie Orthopédique, Hôpital Raymond Poincaré, APHP, Garches, France
| | - Alix Verrando
- Service de Médecine Physique et Réadaptation, HIA Percy, Clamart, F-92140, France
| | - Claire Verdaguer
- Service de Médecine Physique et Réadaptation, HIA Percy, Clamart, F-92140, France
| | - Alain Moussu
- Service de Médecine Physique et Réadaptation, HIA Percy, Clamart, F-92140, France
| | - Aliénor Jequier
- Service de Médecine Physique et Réadaptation, HIA Percy, Clamart, F-92140, France
| | - Christophe Duret
- CRF Les Trois Soleils, Médecine Physique et de Réadaptation, Unité de Rééducation Neurologique, Boissise-Le-Roi, France
| | - Laurence Mailhan
- Service de Médecine Physique et Réadaptation, HIA Percy, Clamart, F-92140, France
| | - Laure Gatin
- CRF Les Trois Soleils, Médecine Physique et de Réadaptation, Unité de Rééducation Neurologique, Boissise-Le-Roi, France
- Service de Chirurgie Orthopédique, Hôpital Raymond Poincaré, APHP, Garches, France
| | - Laurent Oudre
- Université Paris Saclay, Université Paris Cité, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, Gif-sur-Yvette, F-91190, France
| | - Damien Ricard
- Université Paris Cité, Université Paris Saclay, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, Paris, France.
- Service de Neurologie, Service de Santé des Armées, HIA Percy, 101 Avenue Henri Barbusse, Clamart, F-92140, France.
- École du Val-de-Grâce, Service de Santé des Armées, Paris, F-75005, France.
| |
Collapse
|
2
|
Vannelli S, Visintin F, Dosi C, Fiorini L, Rovini E, Cavallo F. A Framework for the Human-Centered Design of Service Processes Enabled by Medical Devices: A Case Study of Wearable Devices for Parkinson's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1367. [PMID: 39457340 PMCID: PMC11507211 DOI: 10.3390/ijerph21101367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
The successful introduction of medical devices (MDs) in real-world settings hinges on designing service processes that cater to stakeholders' needs. While human-centered design (HCD) approaches have been widely applied to service process innovation, the literature lacks a methodology that leverages MDs' key features to design service processes that meet stakeholders' needs. This study aims to fill this gap by developing a framework for the HCD of service processes enabled by MDs. The proposed framework mixes and adapts methodological elements from HCD and technology-enabled design approaches and proposes four new tools. The five-phase framework was applied to the design of a new Parkinson's disease diagnosis and treatment process (PD-DTP) enabled by two wearable MDs for the detection of motor symptoms. The case study lasted five months and involved 42 stakeholders in 21 meetings (interviews, focus groups, etc.). Thanks to the case study, the framework was tested, and a new PD-DTP that could benefit all stakeholders involved was identified. This study provides a framework that, in addition to contributing to theory, could assist MDs developers and healthcare managers in designing service processes that cater to stakeholders' needs by leveraging MDs' key features.
Collapse
Affiliation(s)
- Sara Vannelli
- Dipartimento di Ingegneria Industriale, University of Florence, Viale Morgagni 40/44, 50134 Florence, Italy; (F.V.); (L.F.); (E.R.); (F.C.)
| | - Filippo Visintin
- Dipartimento di Ingegneria Industriale, University of Florence, Viale Morgagni 40/44, 50134 Florence, Italy; (F.V.); (L.F.); (E.R.); (F.C.)
| | - Clio Dosi
- Dipartimento di Scienze Aziendali, University of Bologna, Via Capo di Lucca 34, 40126 Bologna, Italy;
| | - Laura Fiorini
- Dipartimento di Ingegneria Industriale, University of Florence, Viale Morgagni 40/44, 50134 Florence, Italy; (F.V.); (L.F.); (E.R.); (F.C.)
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Viale Rinaldo Piaggio 34, Pontedera, 56025 Pisa, Italy
| | - Erika Rovini
- Dipartimento di Ingegneria Industriale, University of Florence, Viale Morgagni 40/44, 50134 Florence, Italy; (F.V.); (L.F.); (E.R.); (F.C.)
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Viale Rinaldo Piaggio 34, Pontedera, 56025 Pisa, Italy
| | - Filippo Cavallo
- Dipartimento di Ingegneria Industriale, University of Florence, Viale Morgagni 40/44, 50134 Florence, Italy; (F.V.); (L.F.); (E.R.); (F.C.)
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Viale Rinaldo Piaggio 34, Pontedera, 56025 Pisa, Italy
| |
Collapse
|
3
|
Barcellos I, Hansen C, Strobel GK, Geritz J, Munhoz RP, Moscovich M, Maetzler W, Teive HAG. Spatiotemporal Gait Analysis of Patients with Spinocerebellar Ataxia Types 3 and 10 Using Inertial Measurement Units: A Comparative Study. CEREBELLUM (LONDON, ENGLAND) 2024; 23:2109-2121. [PMID: 38869768 DOI: 10.1007/s12311-024-01709-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/03/2024] [Indexed: 06/14/2024]
Abstract
Given the high morbidity related to the progression of gait deficits in spinocerebellar ataxias (SCA), there is a growing interest in identifying biomarkers that can guide early diagnosis and rehabilitation. Spatiotemporal parameter (STP) gait analysis using inertial measurement units (IMUs) has been increasingly studied in this context. This study evaluated STP profiles in SCA types 3 and 10, compared them to controls, and correlated them with clinical scales. IMU portable sensors were used to measure STPs under four gait conditions: self-selected pace (SSP), fast pace (FP), fast pace checking-boxes (FPCB), and fast pace with serial seven subtractions (FPS7). Compared to healthy subjects, both SCA groups had higher values for step time, variability, and swing time, with lower values for gait speed, cadence, and step length. We also found a reduction in speed gain capacity in both SCA groups compared to controls and an increase in speed dual-task cost in the SCA10 group. However, there were no significant differences between the SCA groups. Swing time, mean speed, and step length were correlated with disease severity, risk of falling and functionality in both clinical groups. In the SCA3 group, fear of falling was correlated with cadence. In the SCA10 group, results of the Montreal cognitive assessment test were correlated with step time, mean speed, and step length. These results show that individuals with SCA3 and SCA10 present a highly variable, short-stepped, slow gait pattern compared to healthy subjects, and their gait quality worsened with a fast pace and dual-task involvement.
Collapse
Affiliation(s)
- Igor Barcellos
- Movement Disorders Unit, Neurology Service, Internal Medicine Department, Hospital de Clínicas, Federal University of Paraná, Curitiba, Brazil.
| | - Clint Hansen
- Department of Neurology, University Hospital Schleswig-Holstein and Kiel University, Kiel, Germany
| | - Giovanna Klüppel Strobel
- Movement Disorders Unit, Neurology Service, Internal Medicine Department, Hospital de Clínicas, Federal University of Paraná, Curitiba, Brazil
| | - Johanna Geritz
- Department of Neurology, University Hospital Schleswig-Holstein and Kiel University, Kiel, Germany
| | - Renato P Munhoz
- Gloria and Morton Shulman Movement Disorders Centre, Toronto Western Hospital, University of Toronto, Toronto, Canada
| | - Mariana Moscovich
- Movement Disorders Unit, Neurology Service, Internal Medicine Department, Hospital de Clínicas, Federal University of Paraná, Curitiba, Brazil
| | - Walter Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein and Kiel University, Kiel, Germany
| | - Hélio Afonso Ghizoni Teive
- Movement Disorders Unit, Neurology Service, Internal Medicine Department, Hospital de Clínicas, Federal University of Paraná, Curitiba, Brazil
| |
Collapse
|
4
|
Choi H, Youm C, Park H, Kim B, Hwang J, Cheon SM, Shin S. Convolutional neural network based detection of early stage Parkinson's disease using the six minute walk test. Sci Rep 2024; 14:22648. [PMID: 39349539 PMCID: PMC11442580 DOI: 10.1038/s41598-024-72648-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/09/2024] [Indexed: 10/02/2024] Open
Abstract
The heterogeneity of Parkinson's disease (PD) presents considerable challenges for accurate diagnosis, particularly during early-stage disease, when the symptoms may be extremely subtle. This study aimed to assess the accuracy of a convolutional neural network (CNN) technique based on the 6-min walk test (6MWT) measured using wearable sensors to distinguish patients with early-stage PD (n = 78) from healthy controls (n = 50). The participants wore six sensors, and performed the 6MWT. The time-series data were converted into new images. The results revealed that the gyroscopic vertical component of the lumbar spine displayed the highest classification accuracy of 83.5%, followed by those of the thoracic spine (83.1%) and right thigh (79.5%) segment. These findings suggest that the 6MWT and CNN models may facilitate earlier diagnosis and monitoring of PD symptoms, enabling clinicians to provide timely treatment during the critical transition from normal to pathologic gait patterns.
Collapse
Affiliation(s)
- Hyejin Choi
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Republic of Korea
| | - Changhong Youm
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Republic of Korea.
| | - Hwayoung Park
- Biomechanics Laboratory, Dong-A University, Busan, Republic of Korea
| | - Bohyun Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Republic of Korea
| | - Juseon Hwang
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Republic of Korea
| | - Sang-Myung Cheon
- Department of Neurology, School of Medicine, Dong-A University, Busan, Republic of Korea
| | - Sungtae Shin
- Department of Mechanical Engineering, College of Engineering, Dong-A University, Busan, Republic of Korea
| |
Collapse
|
5
|
Oppermann J, Tschentscher V, Welzel J, Geritz J, Hansen C, Gold R, Maetzler W, Scherbaum R, Tönges L. Clinical and device-based predictors of improved experience of activities of daily living after a multidisciplinary inpatient treatment for people with Parkinson's disease: a cohort study. Ther Adv Neurol Disord 2024; 17:17562864241277157. [PMID: 39328922 PMCID: PMC11425784 DOI: 10.1177/17562864241277157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 08/01/2024] [Indexed: 09/28/2024] Open
Abstract
Background The inpatient Parkinson's Disease Multimodal Complex Treatment (PD-MCT) is an important therapeutical approach to improving gait and activities of daily living (ADL) of people with PD (PwP). Wearable device-based parameters (DBP) are new options for specific gait analyses toward individualized treatments. Objectives We sought to identify predictors of perceived ADL benefit taking clinical scores and DBP into account. Additionally, we analyzed DBP and clinical scores before and after PD-MCT. Design Exploratory observational cohort study. Methods Clinical scores and DBP of 56 PwP (mean age: 66.3 years, median Hoehn and Yahr (H&Y) stage: 2.5) were examined at the start and the end of a 14-day inpatient PD-MCT in a German University Medical Center. Participants performed four straight walking tasks under single- and dual-task conditions for gait analyses. Additionally, clinical scores of motor and nonmotor functions and quality of life (QoL) were assessed. Using dichotomized data of change in Movement Disorders Society Unified Parkinson's Disease Rating Scale Part II (MDS-UPDRS II) as a dependent variable and clinical and DBP as independent variables, a binomial logistic regression model was implemented. Results Young age, high perceived ADL impairment at baseline, high dexterity skills, and a steady gait were significant predictors of ADL benefit after PD-MCT. DBP like gait speed, number of steps, step time, stance time, and double limb support time were improved after PD-MCT. In addition, motor functions (e.g., MDS-UPDRS III and IV), QoL, perceived ADL (MDS-UPDRS II), and experience of nonmotor functions (MDS-UPDRS I) improved significantly. Conclusion The logistic regression model identified a group of PwP who had the most probable perceived ADL benefit after PD-MCT. Additionally, gait improved toward a faster and more dynamic gait. Using wearable technology in context of PD-MCT is promising to offer more personalized therapeutical concepts. Trial registration German Clinical Trial Register, https://drks.de; DRKS00020948 number, 30 March 2020, retrospectively registered.
Collapse
Affiliation(s)
- Judith Oppermann
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Vera Tschentscher
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Julius Welzel
- Department of Neurology, Kiel University, Kiel, Germany
| | | | - Clint Hansen
- Department of Neurology, Kiel University, Kiel, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
- Neurodegeneration Research, Protein Research Unit Ruhr (PURE), Ruhr University Bochum, Bochum, Germany
| | | | - Raphael Scherbaum
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, Gudrunstr. 56, Bochum D-44791, Germany
| | - Lars Tönges
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
- Neurodegeneration Research, Protein Research Unit Ruhr (PURE), Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
6
|
Khalil RM, Shulman LM, Gruber-Baldini AL, Shakya S, Fenderson R, Van Hoven M, Hausdorff JM, von Coelln R, Cummings MP. Simplification of Mobility Tests and Data Processing to Increase Applicability of Wearable Sensors as Diagnostic Tools for Parkinson's Disease. SENSORS (BASEL, SWITZERLAND) 2024; 24:4983. [PMID: 39124030 PMCID: PMC11314738 DOI: 10.3390/s24154983] [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] [Received: 06/19/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
Quantitative mobility analysis using wearable sensors, while promising as a diagnostic tool for Parkinson's disease (PD), is not commonly applied in clinical settings. Major obstacles include uncertainty regarding the best protocol for instrumented mobility testing and subsequent data processing, as well as the added workload and complexity of this multi-step process. To simplify sensor-based mobility testing in diagnosing PD, we analyzed data from 262 PD participants and 50 controls performing several motor tasks wearing a sensor on their lower back containing a triaxial accelerometer and a triaxial gyroscope. Using ensembles of heterogeneous machine learning models incorporating a range of classifiers trained on a set of sensor features, we show that our models effectively differentiate between participants with PD and controls, both for mixed-stage PD (92.6% accuracy) and a group selected for mild PD only (89.4% accuracy). Omitting algorithmic segmentation of complex mobility tasks decreased the diagnostic accuracy of our models, as did the inclusion of kinesiological features. Feature importance analysis revealed that Timed Up and Go (TUG) tasks to contribute the highest-yield predictive features, with only minor decreases in accuracy for models based on cognitive TUG as a single mobility task. Our machine learning approach facilitates major simplification of instrumented mobility testing without compromising predictive performance.
Collapse
Affiliation(s)
- Rana M. Khalil
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA;
| | - Lisa M. Shulman
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (L.M.S.); (R.F.); (M.V.H.)
| | - Ann L. Gruber-Baldini
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.L.G.-B.); (S.S.)
| | - Sunita Shakya
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.L.G.-B.); (S.S.)
| | - Rebecca Fenderson
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (L.M.S.); (R.F.); (M.V.H.)
| | - Maxwell Van Hoven
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (L.M.S.); (R.F.); (M.V.H.)
| | - Jeffrey M. Hausdorff
- Center for the Study of Movement, Cognition, and Mobility, Neurological Institute, Tel Aviv Medical Center, Tel Aviv 6492416, Israel;
- Department of Physical Therapy, Faculty of Medicine & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Rainer von Coelln
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (L.M.S.); (R.F.); (M.V.H.)
| | - Michael P. Cummings
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742, USA;
| |
Collapse
|
7
|
Barbosa R, Mendonça M, Bastos P, Pita Lobo P, Valadas A, Correia Guedes L, Ferreira JJ, Rosa MM, Matias R, Coelho M. 3D Kinematics Quantifies Gait Response to Levodopa earlier and to a more Comprehensive Extent than the MDS-Unified Parkinson's Disease Rating Scale in Patients with Motor Complications. Mov Disord Clin Pract 2024; 11:795-807. [PMID: 38610081 PMCID: PMC11233852 DOI: 10.1002/mdc3.14016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/20/2024] [Accepted: 02/13/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Quantitative 3D movement analysis using inertial measurement units (IMUs) allows for a more detailed characterization of motor patterns than clinical assessment alone. It is essential to discriminate between gait features that are responsive or unresponsive to current therapies to better understand the underlying pathophysiological basis and identify potential therapeutic strategies. OBJECTIVES This study aims to characterize the responsiveness and temporal evolution of different gait subcomponents in Parkinson's disease (PD) patients in their OFF and various ON states following levodopa administration, utilizing both wearable sensors and the gold-standard MDS-UPDRS motor part III. METHODS Seventeen PD patients were assessed while wearing a full-body set of 15 IMUs in their OFF state and at 20-minute intervals following the administration of a supra-threshold levodopa dose. Gait was reconstructed using a biomechanical model of the human body to quantify how each feature was modulated. Comparisons with non-PD control subjects were conducted in parallel. RESULTS Significant motor changes were observed in both the upper and lower limbs according to the MDS-UPDRS III, 40 minutes after levodopa intake. IMU-assisted 3D kinematics detected significant motor alterations as early as 20 minutes after levodopa administration, particularly in upper limbs metrics. Although all "pace-domain" gait features showed significant improvement in the Best-ON state, most rhythmicity, asymmetry, and variability features did not. CONCLUSION IMUs are capable of detecting motor alterations earlier and in a more comprehensive manner than the MDS-UPDRS III. The upper limbs respond more rapidly to levodopa, possibly reflecting distinct thresholds to levodopa across striatal regions.
Collapse
Affiliation(s)
- Raquel Barbosa
- Neurology DeparmentCentre Hospitalier Universitaire ToulouseToulouseFrance
- Nova Medical School, Faculdade de Ciências MedicasUniversidade Nova de LisboaLisbonPortugal
| | - Marcelo Mendonça
- Nova Medical School, Faculdade de Ciências MedicasUniversidade Nova de LisboaLisbonPortugal
- Champalimaud Research and Clinical Centre, Champalimaud Centre for the UnknownLisbonPortugal
| | - Paulo Bastos
- Neurology DeparmentCentre Hospitalier Universitaire ToulouseToulouseFrance
- Nova Medical School, Faculdade de Ciências MedicasUniversidade Nova de LisboaLisbonPortugal
| | - Patrícia Pita Lobo
- Department of Neurosciences and Mental HealthNeurology Hospital Santa Maria, CHLUNLisbonPortugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of MedicineUniversity of LisbonLisbonPortugal
| | - Anabela Valadas
- Department of Neurosciences and Mental HealthNeurology Hospital Santa Maria, CHLUNLisbonPortugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of MedicineUniversity of LisbonLisbonPortugal
| | - Leonor Correia Guedes
- Department of Neurosciences and Mental HealthNeurology Hospital Santa Maria, CHLUNLisbonPortugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of MedicineUniversity of LisbonLisbonPortugal
| | - Joaquim J. Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of MedicineUniversity of LisbonLisbonPortugal
- Laboratory of Clinical Pharmacology and Therapeutics, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
- CNS‐ Campus Neurológico SeniorTorres VedrasPortugal
| | - Mário Miguel Rosa
- Department of Neurosciences and Mental HealthNeurology Hospital Santa Maria, CHLUNLisbonPortugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of MedicineUniversity of LisbonLisbonPortugal
- Laboratory of Clinical Pharmacology and Therapeutics, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
| | - Ricardo Matias
- Physics Department & Institute of Biophysics and Biomedical Engineering (IBEB), Faculty of SciencesUniversity of LisbonLisbonPortugal
- KinetikosCoimbraPortugal
| | - Miguel Coelho
- Department of Neurosciences and Mental HealthNeurology Hospital Santa Maria, CHLUNLisbonPortugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculty of MedicineUniversity of LisbonLisbonPortugal
| |
Collapse
|
8
|
Adams JL, Kangarloo T, Gong Y, Khachadourian V, Tracey B, Volfson D, Latzman RD, Cosman J, Edgerton J, Anderson D, Best A, Kostrzebski MA, Auinger P, Wilmot P, Pohlson Y, Jensen-Roberts S, Müller MLTM, Stephenson D, Dorsey ER. Using a smartwatch and smartphone to assess early Parkinson's disease in the WATCH-PD study over 12 months. NPJ Parkinsons Dis 2024; 10:112. [PMID: 38866793 PMCID: PMC11169239 DOI: 10.1038/s41531-024-00721-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/10/2024] [Indexed: 06/14/2024] Open
Abstract
Digital measures may provide objective, sensitive, real-world measures of disease progression in Parkinson's disease (PD). However, multicenter longitudinal assessments of such measures are few. We recently demonstrated that baseline assessments of gait, tremor, finger tapping, and speech from a commercially available smartwatch, smartphone, and research-grade wearable sensors differed significantly between 82 individuals with early, untreated PD and 50 age-matched controls. Here, we evaluated the longitudinal change in these assessments over 12 months in a multicenter observational study using a generalized additive model, which permitted flexible modeling of at-home data. All measurements were included until participants started medications for PD. Over one year, individuals with early PD experienced significant declines in several measures of gait, an increase in the proportion of day with tremor, modest changes in speech, and few changes in psychomotor function. As measured by the smartwatch, the average (SD) arm swing in-clinic decreased from 25.9 (15.3) degrees at baseline to 19.9 degrees (13.7) at month 12 (P = 0.004). The proportion of awake time an individual with early PD had tremor increased from 19.3% (18.0%) to 25.6% (21.4%; P < 0.001). Activity, as measured by the number of steps taken per day, decreased from 3052 (1306) steps per day to 2331 (2010; P = 0.16), but this analysis was restricted to 10 participants due to the exclusion of those that had started PD medications and lost the data. The change of these digital measures over 12 months was generally larger than the corresponding change in individual items on the Movement Disorder Society-Unified Parkinson's Disease Rating Scale but not greater than the change in the overall scale. Successful implementation of digital measures in future clinical trials will require improvements in study conduct, especially data capture. Nonetheless, gait and tremor measures derived from a commercially available smartwatch and smartphone hold promise for assessing the efficacy of therapeutics in early PD.
Collapse
Affiliation(s)
- Jamie L Adams
- Center for Health + Technology, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA.
| | | | - Yishu Gong
- Takeda Pharmaceuticals, Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | - Melissa A Kostrzebski
- Center for Health + Technology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Peggy Auinger
- Center for Health + Technology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Peter Wilmot
- Center for Health + Technology, University of Rochester Medical Center, Rochester, NY, USA
| | - Yvonne Pohlson
- Center for Health + Technology, University of Rochester Medical Center, Rochester, NY, USA
| | - Stella Jensen-Roberts
- Center for Health + Technology, University of Rochester Medical Center, Rochester, NY, USA
| | | | | | - E Ray Dorsey
- Center for Health + Technology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
9
|
Zhang X, Jin Y, Wang M, Ji C, Chen Z, Fan W, Rainer TH, Guan Q, Li Q. The impact of anxiety on gait impairments in Parkinson's disease: insights from sensor-based gait analysis. J Neuroeng Rehabil 2024; 21:68. [PMID: 38689288 PMCID: PMC11059709 DOI: 10.1186/s12984-024-01364-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 04/22/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Sensor-based gait analysis provides a robust quantitative tool for assessing gait impairments and their associated factors in Parkinson's disease (PD). Anxiety is observed to interfere with gait clinically, but this has been poorly investigated. Our purpose is to utilize gait analysis to uncover the effect of anxiety on gait in patients with PD. METHODS We enrolled 38 and 106 PD patients with and without anxiety, respectively. Gait parameters were quantitively examined and compared between two groups both in single-task (ST) and dual-task (DT) walking tests. Multiple linear regression was applied to evaluate whether anxiety independently contributed to gait impairments. RESULTS During ST, PD patients with anxiety presented significantly shorter stride length, lower gait velocity, longer stride time and stance time, longer stance phase, smaller toe-off (TO) and heel-strike (HS) angles than those without anxiety. While under DT status, the differences were diminished. Multiple linear regression analysis demonstrated that anxiety was an independent factor to a serials of gait parameters, particularly ST-TO (B = -2.599, (-4.82, -0.38)), ST-HS (B = -2.532, (-4.71, -0.35)), ST-TO-CV (B = 4.627, (1.71, 7.64)), ST-HS-CV(B = 4.597, (1.66, 7.53)), ST stance phase (B = 1.4, (0.22, 2.58)), and DT stance phase (B = 1.749, (0.56, 2.94)). CONCLUSION Our study discovered that anxiety has a significant impact on gait impairments in PD patients, especially exacerbating shuffling steps and prolonging stance phase. These findings highlight the importance of addressing anxiety in PD precision therapy to achieve better treatment outcomes.
Collapse
Affiliation(s)
- Xiaodan Zhang
- Department of Neurology, Ningbo NO.2 Hospital, Ningbo, Zhejiang Province, China
- Department of Emergency Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Yulan Jin
- Department of Clinical Laboratory, Ningbo NO.2 Hospital, Ningbo, Zhejiang Province, China
| | - Mateng Wang
- Department of General Surgery, Yinzhou NO.2 Hospital, Ningbo, Zhejiang Province, China
| | - Chengcheng Ji
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Zhaoying Chen
- Department of Neurology, Ningbo NO.2 Hospital, Ningbo, Zhejiang Province, China
| | - Weinv Fan
- Department of Neurology, Ningbo NO.2 Hospital, Ningbo, Zhejiang Province, China
| | | | - Qiongfeng Guan
- Department of Neurology, Ningbo NO.2 Hospital, Ningbo, Zhejiang Province, China.
| | - Qianyun Li
- Department of Neurology, Ningbo NO.2 Hospital, Ningbo, Zhejiang Province, China.
- Department of Emergency Medicine, University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
10
|
Boborzi L, Decker J, Rezaei R, Schniepp R, Wuehr M. Human Activity Recognition in a Free-Living Environment Using an Ear-Worn Motion Sensor. SENSORS (BASEL, SWITZERLAND) 2024; 24:2665. [PMID: 38732771 PMCID: PMC11085719 DOI: 10.3390/s24092665] [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] [Received: 03/29/2024] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024]
Abstract
Human activity recognition (HAR) technology enables continuous behavior monitoring, which is particularly valuable in healthcare. This study investigates the viability of using an ear-worn motion sensor for classifying daily activities, including lying, sitting/standing, walking, ascending stairs, descending stairs, and running. Fifty healthy participants (between 20 and 47 years old) engaged in these activities while under monitoring. Various machine learning algorithms, ranging from interpretable shallow models to state-of-the-art deep learning approaches designed for HAR (i.e., DeepConvLSTM and ConvTransformer), were employed for classification. The results demonstrate the ear sensor's efficacy, with deep learning models achieving a 98% accuracy rate of classification. The obtained classification models are agnostic regarding which ear the sensor is worn and robust against moderate variations in sensor orientation (e.g., due to differences in auricle anatomy), meaning no initial calibration of the sensor orientation is required. The study underscores the ear's efficacy as a suitable site for monitoring human daily activity and suggests its potential for combining HAR with in-ear vital sign monitoring. This approach offers a practical method for comprehensive health monitoring by integrating sensors in a single anatomical location. This integration facilitates individualized health assessments, with potential applications in tele-monitoring, personalized health insights, and optimizing athletic training regimes.
Collapse
Affiliation(s)
- Lukas Boborzi
- German Center for Vertigo and Balance Disorders (DSGZ), Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
| | - Julian Decker
- German Center for Vertigo and Balance Disorders (DSGZ), Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
| | - Razieh Rezaei
- German Center for Vertigo and Balance Disorders (DSGZ), Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
| | - Roman Schniepp
- Institute for Emergency Medicine and Medical Management, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany
| | - Max Wuehr
- German Center for Vertigo and Balance Disorders (DSGZ), Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
- Department of Neurology, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany
| |
Collapse
|
11
|
Moorthy P, Weinert L, Schüttler C, Svensson L, Sedlmayr B, Müller J, Nagel T. Attributes, Methods, and Frameworks Used to Evaluate Wearables and Their Companion mHealth Apps: Scoping Review. JMIR Mhealth Uhealth 2024; 12:e52179. [PMID: 38578671 PMCID: PMC11031706 DOI: 10.2196/52179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/15/2023] [Accepted: 02/01/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Wearable devices, mobile technologies, and their combination have been accepted into clinical use to better assess the physical fitness and quality of life of patients and as preventive measures. Usability is pivotal for overcoming constraints and gaining users' acceptance of technology such as wearables and their companion mobile health (mHealth) apps. However, owing to limitations in design and evaluation, interactive wearables and mHealth apps have often been restricted from their full potential. OBJECTIVE This study aims to identify studies that have incorporated wearable devices and determine their frequency of use in conjunction with mHealth apps or their combination. Specifically, this study aims to understand the attributes and evaluation techniques used to evaluate usability in the health care domain for these technologies and their combinations. METHODS We conducted an extensive search across 4 electronic databases, spanning the last 30 years up to December 2021. Studies including the keywords "wearable devices," "mobile apps," "mHealth apps," "physiological data," "usability," "user experience," and "user evaluation" were considered for inclusion. A team of 5 reviewers screened the collected publications and charted the features based on the research questions. Subsequently, we categorized these characteristics following existing usability and wearable taxonomies. We applied a methodological framework for scoping reviews and the PRISMA-ScR (Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews) checklist. RESULTS A total of 382 reports were identified from the search strategy, and 68 articles were included. Most of the studies (57/68, 84%) involved the simultaneous use of wearables and connected mobile apps. Wrist-worn commercial consumer devices such as wristbands were the most prevalent, accounting for 66% (45/68) of the wearables identified in our review. Approximately half of the data from the medical domain (32/68, 47%) focused on studies involving participants with chronic illnesses or disorders. Overall, 29 usability attributes were identified, and 5 attributes were frequently used for evaluation: satisfaction (34/68, 50%), ease of use (27/68, 40%), user experience (16/68, 24%), perceived usefulness (18/68, 26%), and effectiveness (15/68, 22%). Only 10% (7/68) of the studies used a user- or human-centered design paradigm for usability evaluation. CONCLUSIONS Our scoping review identified the types and categories of wearable devices and mHealth apps, their frequency of use in studies, and their implementation in the medical context. In addition, we examined the usability evaluation of these technologies: methods, attributes, and frameworks. Within the array of available wearables and mHealth apps, health care providers encounter the challenge of selecting devices and companion apps that are effective, user-friendly, and compatible with user interactions. The current gap in usability and user experience in health care research limits our understanding of the strengths and limitations of wearable technologies and their companion apps. Additional research is necessary to overcome these limitations.
Collapse
Affiliation(s)
- Preetha Moorthy
- Department of Biomedical Informatics, Center for Preventive Medicine and Digital Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lina Weinert
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
- Section for Oral Health, Heidelberg Institute of Global Health, Heidelberg University Hospital, Heidelberg, Germany
| | - Christina Schüttler
- Medical Center for Information and Communication Technology, University Hospital Erlangen, Erlangen, Germany
| | - Laura Svensson
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Brita Sedlmayr
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Julia Müller
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Till Nagel
- Human Data Interaction Lab, Mannheim University of Applied Sciences, Mannheim, Germany
| |
Collapse
|
12
|
Zatti C, Pilotto A, Hansen C, Rizzardi A, Catania M, Romijnders R, Purin L, Pasolini MP, Schaeffer E, Galbiati A, Ferini-Strambi L, Berg D, Maetzler W, Padovani A. Turning alterations detected by mobile health technology in idiopathic REM sleep behavior disorder. NPJ Parkinsons Dis 2024; 10:64. [PMID: 38499543 PMCID: PMC10948811 DOI: 10.1038/s41531-024-00682-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 03/12/2024] [Indexed: 03/20/2024] Open
Abstract
Idiopathic REM sleep Behavior Disorder (iRBD) is a condition at high risk of developing Parkinson's disease (PD) and other alpha-synucleinopathies. The aim of the study was to evaluate subtle turning alterations by using Mobile health technology in iRBD individuals without subthreshold parkinsonism. A total of 148 participants (23 persons with polysomnography-confirmed iRBD without subthreshold parkinsonism, 60 drug-naïve PD patients, and 65 age-matched controls were included in this prospective cross-sectional study. All underwent a multidimensional assessment including cognitive and non-motor symptoms assessment. Then a Timed-Up-and-Go test (TUG) at normal and fast speed was performed using mobile health technology on the lower back (Rehagait®, Hasomed, Germany). Duration, mean, and peak angular velocities of the turns were compared using a multivariate model correcting for age and sex. Compared to controls, PD patients showed longer turn durations and lower mean and peak angular velocities of the turns in both TUGs (all p ≤ 0.001). iRBD participants also showed a longer turn duration and lower mean (p = 0.006) and peak angular velocities (p < 0.001) compared to controls, but only in the TUG at normal speed. Mobile health technology assessment identified subtle alterations of turning in subjects with iRBD in usual, but not fast speed. Longitudinal studies are warranted to evaluate the value of objective turning parameters in defining the risk of conversion to PD in iRBD and in tracking motor progression in prodromal PD.
Collapse
Affiliation(s)
- Cinzia Zatti
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Laboratory of digital Neurology and biosensors, University of Brescia, Brescia, Italy
- Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Andrea Pilotto
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy.
- Laboratory of digital Neurology and biosensors, University of Brescia, Brescia, Italy.
- Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili of Brescia, Brescia, Italy.
| | - Clint Hansen
- Department of Neurology, University Hospital Schleswig-Holstein and Kiel University, Kiel, Germany
| | - Andrea Rizzardi
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Laboratory of digital Neurology and biosensors, University of Brescia, Brescia, Italy
| | - Marcello Catania
- Laboratory of digital Neurology and biosensors, University of Brescia, Brescia, Italy
- Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Robbin Romijnders
- Department of Neurology, University Hospital Schleswig-Holstein and Kiel University, Kiel, Germany
| | - Leandro Purin
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Laboratory of digital Neurology and biosensors, University of Brescia, Brescia, Italy
- Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Maria P Pasolini
- Department of Clinical and Experimental Sciences, Neurophysiology Unit, University of Brescia, Brescia, Italy
| | - Eva Schaeffer
- Department of Neurology, University Hospital Schleswig-Holstein and Kiel University, Kiel, Germany
| | - Andrea Galbiati
- IRCCS San Raffaele Scientific Institute, Department of Clinical Neurosciences, Neurology-Sleep Disorders Centre, Milan, Italy
- Faculty of Psychology, "Vita-Salute" San Raffaele University, Milan, Italy
| | - Luigi Ferini-Strambi
- IRCCS San Raffaele Scientific Institute, Department of Clinical Neurosciences, Neurology-Sleep Disorders Centre, Milan, Italy
- Faculty of Psychology, "Vita-Salute" San Raffaele University, Milan, Italy
| | - Daniela Berg
- Department of Neurology, University Hospital Schleswig-Holstein and Kiel University, Kiel, Germany
| | - Walter Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein and Kiel University, Kiel, Germany
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Laboratory of digital Neurology and biosensors, University of Brescia, Brescia, Italy
- Department of continuity of care and frailty, Neurology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
- Department of Clinical and Experimental Sciences, Neurophysiology Unit, University of Brescia, Brescia, Italy
| |
Collapse
|
13
|
Huang J, Lin L, Yu F, He X, Song W, Lin J, Tang Z, Yuan K, Li Y, Huang H, Pei Z, Xian W, Yu-Chian Chen C. Parkinson's severity diagnosis explainable model based on 3D multi-head attention residual network. Comput Biol Med 2024; 170:107959. [PMID: 38215619 DOI: 10.1016/j.compbiomed.2024.107959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/31/2023] [Accepted: 01/01/2024] [Indexed: 01/14/2024]
Abstract
The severity evaluation of Parkinson's disease (PD) is of great significance for the treatment of PD. However, existing methods either have limitations based on prior knowledge or are invasive methods. To propose a more generalized severity evaluation model, this paper proposes an explainable 3D multi-head attention residual convolution network. First, we introduce the 3D attention-based convolution layer to extract video features. Second, features will be fed into LSTM and residual backbone networks, which can be used to capture the contextual information of the video. Finally, we design a feature compression module to condense the learned contextual features. We develop some interpretable experiments to better explain this black-box model so that it can be better generalized. Experiments show that our model can achieve state-of-the-art diagnosis performance. The proposed lightweight but effective model is expected to serve as a suitable end-to-end deep learning baseline in future research on PD video-based severity evaluation and has the potential for large-scale application in PD telemedicine. The source code is available at https://github.com/JackAILab/MARNet.
Collapse
Affiliation(s)
- Jiehui Huang
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Lishan Lin
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, 510080, China
| | - Fengcheng Yu
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Xuedong He
- School of Computer Science and Technology, Zhejiang Normal University, Jinhua, 321004, Zhejiang, China
| | - Wenhui Song
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Jiaying Lin
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Zhenchao Tang
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Kang Yuan
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, 510080, China
| | - Yucheng Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, 510080, China
| | - Haofan Huang
- Polytechnic Institute, Zhejiang University, Hangzhou 310058, China
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, 510080, China.
| | - Wenbiao Xian
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, 510080, China.
| | - Calvin Yu-Chian Chen
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China; AI for Science (AI4S)-Preferred Program, Peking University Shenzhen Graduate School, Shenzhen, 518055, Guangdong, China; School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, Guangdong, China; Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan; Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan.
| |
Collapse
|
14
|
Baudendistel ST, Haussler AM, Rawson KS, Earhart GM. Minimal clinically important differences of spatiotemporal gait variables in Parkinson disease. Gait Posture 2024; 108:257-263. [PMID: 38150946 PMCID: PMC10878409 DOI: 10.1016/j.gaitpost.2023.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/18/2023] [Accepted: 11/21/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Assessment of gait function in People with Parkinson Disease (PwPD) is an important tool for monitoring disease progression in PD. While comprehensive gait analysis has become increasingly popular, only one study, Hass et al. (2014), has established minimal clinically important differences (MCID) for one spatiotemporal variable (velocity) in PwPD. RESEARCH QUESTION What are the MCIDs for velocity and additional spatiotemporal variables, including mean, variability, and asymmetry of step length, time, and width? METHODS As part of a larger clinic-based initiative, 382 medicated, ambulatory PwPD walked on an instrumented walkway during routine clinical visits. Distribution and anchor-based methods (Unified Parkinson's Disease Rating Scale-III, Modified Hoehn and Yahr, and the mobility subsection of the Parkinson Disease Questionnaire) were used to calculate MCIDs for variables of interest in a cross-sectional approach. RESULTS Distribution measures for all variables are presented. Of nine gait variables, four were significantly associated with every anchor and pooled to the following values: velocity (8.2 cm/s), step length mean (3.6 cm), step length variability (0.7%), and step time variability (0.67%). SIGNIFICANCE The finalized MCID for velocity (8.2 cm/s) was nearly half of the MCID of 15 cm/s reported by Hass et al., potentially due to differences in calculations. These results allow for evaluations of effectiveness of interventions by providing values that are specific to changes in gait for PwPD. Alterations of methodology including different versions of clinical or walking assessments, and/or different calculation and selection of gait variables necessitate careful reasoning when using presented MCIDs.
Collapse
Affiliation(s)
- Sidney T Baudendistel
- Program in Physical Therapy, Washington University School of Medicine in St. Louis, United States
| | - Allison M Haussler
- Program in Physical Therapy, Washington University School of Medicine in St. Louis, United States
| | - Kerri S Rawson
- Program in Physical Therapy, Washington University School of Medicine in St. Louis, United States; Department of Neurology, Washington University School of Medicine in St. Louis, United States
| | - Gammon M Earhart
- Program in Physical Therapy, Washington University School of Medicine in St. Louis, United States; Department of Neurology, Washington University School of Medicine in St. Louis, United States; Department of Neuroscience, Washington University School of Medicine in St. Louis, United States.
| |
Collapse
|
15
|
Wang Q, Zeng W, Dai X. Gait classification for early detection and severity rating of Parkinson's disease based on hybrid signal processing and machine learning methods. Cogn Neurodyn 2024; 18:109-132. [PMID: 38406205 PMCID: PMC10881932 DOI: 10.1007/s11571-022-09925-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 12/03/2022] [Accepted: 12/19/2022] [Indexed: 12/31/2022] Open
Abstract
Parkinson's disease (PD) is one of the cognitive degenerative disorders of the central nervous system that affects the motor system. Gait dysfunction represents the pathology of motor symptom while gait analysis provides clinicians with subclinical information reflecting subtle differences between PD patients and healthy controls (HCs). Currently neurologists usually assess several clinical manifestations of the PD patients and rate the severity level according to some established criteria. This is highly dependent on clinician's expertise which is subjective and ineffective. In the present study we address these issues by proposing a hybrid signal processing and machine learning based gait classification system for gait anomaly detection and severity rating of PD patients. Time series of vertical ground reaction force (VGRF) data are utilized to represent discriminant gait information. First, phase space of the VGRF is reconstructed, in which the properties associated with the nonlinear gait system dynamics are preserved. Then Shannon energy is used to extract the characteristic envelope of the phase space signal. Third, Shannon energy envelope is decomposed into high and low resonance components using dual Q-factor signal decomposition derived from tunable Q-factor wavelet transform. Note that the high Q-factor component consists largely of sustained oscillatory behavior, while the low Q-factor component consists largely of transients and oscillations that are not sustained. Fourth, variational mode decomposition is employed to decompose high and low resonance components into different intrinsic modes and provide representative features. Finally features are fed to five different types of machine learning based classifiers for the anomaly detection and severity rating of PD patients based on Hohen and Yahr (HY) scale. The effectiveness of this strategy is verified using a Physionet gait database consisting of 93 idiopathic PD patients and 73 age-matched asymptomatic HCs. When evaluated with 10-fold cross-validation method for early PD detection and severity rating, the highest classification accuracy is reported to be 98.20 % and 96.69 % , respectively, by using the support vector machine classifier. Compared with other state-of-the-art methods, the results demonstrate superior performance and support the validity of the proposed method.
Collapse
Affiliation(s)
- Qinghui Wang
- School of Physics and Mechanical and Electrical Engineering, Longyan University, Longyan, 364012 People's Republic of China
| | - Wei Zeng
- School of Physics and Mechanical and Electrical Engineering, Longyan University, Longyan, 364012 People's Republic of China
| | - Xiangkun Dai
- School of Physics and Mechanical and Electrical Engineering, Longyan University, Longyan, 364012 People's Republic of China
| |
Collapse
|
16
|
Maetzler W, Mirelman A, Pilotto A, Bhidayasiri R. Identifying Subtle Motor Deficits Before Parkinson's Disease is Diagnosed: What to Look for? JOURNAL OF PARKINSON'S DISEASE 2024; 14:S287-S296. [PMID: 38363620 PMCID: PMC11492040 DOI: 10.3233/jpd-230350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/11/2024] [Indexed: 02/17/2024]
Abstract
Motor deficits typical of Parkinson's disease (PD), such as gait and balance disturbances, tremor, reduced arm swing and finger movement, and voice and breathing changes, are believed to manifest several years prior to clinical diagnosis. Here we describe the evidence for the presence and progression of motor deficits in this pre-diagnostic phase in order to provide suggestions for the design of future observational studies for an effective, quantitatively oriented investigation. On the one hand, these future studies must detect these motor deficits in as large (potentially, population-based) cohorts as possible with high sensitivity and specificity. On the other hand, they must describe the progression of these motor deficits in the pre-diagnostic phase as accurately as possible, to support the testing of the effect of pharmacological and non-pharmacological interventions. Digital technologies and artificial intelligence can substantially accelerate this process.
Collapse
Affiliation(s)
- Walter Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein and Kiel University, Kiel, Germany
| | - Anat Mirelman
- Laboratory for Early Markers of Neurodegeneration, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sagol School of Neuroscience and Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Roongroj Bhidayasiri
- Chulalongkorn Centre of Excellence for Parkinson’s Disease & Related Disorders, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| |
Collapse
|
17
|
Burtscher J, Moraud EM, Malatesta D, Millet GP, Bally JF, Patoz A. Exercise and gait/movement analyses in treatment and diagnosis of Parkinson's Disease. Ageing Res Rev 2024; 93:102147. [PMID: 38036102 DOI: 10.1016/j.arr.2023.102147] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/23/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Cardinal motor symptoms in Parkinson's disease (PD) include bradykinesia, rest tremor and/or rigidity. This symptomatology can additionally encompass abnormal gait, balance and postural patterns at advanced stages of the disease. Besides pharmacological and surgical therapies, physical exercise represents an important strategy for the management of these advanced impairments. Traditionally, diagnosis and classification of such abnormalities have relied on partially subjective evaluations performed by neurologists during short and temporally scattered hospital appointments. Emerging sports medical methods, including wearable sensor-based movement assessment and computational-statistical analysis, are paving the way for more objective and systematic diagnoses in everyday life conditions. These approaches hold promise to facilitate customizing clinical trials to specific PD groups, as well as personalizing neuromodulation therapies and exercise prescriptions for each individual, remotely and regularly, according to disease progression or specific motor symptoms. We aim to summarize exercise benefits for PD with a specific emphasis on gait and balance deficits, and to provide an overview of recent advances in movement analysis approaches, notably from the sports science community, with value for diagnosis and prognosis. Although such techniques are becoming increasingly available, their standardization and optimization for clinical purposes is critically missing, especially in their translation to complex neurodegenerative disorders such as PD. We highlight the importance of integrating state-of-the-art gait and movement analysis approaches, in combination with other motor, electrophysiological or neural biomarkers, to improve the understanding of the diversity of PD phenotypes, their response to therapies and the dynamics of their disease progression.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland.
| | - Eduardo Martin Moraud
- Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland; Defitech Centre for Interventional Neurotherapies (NeuroRestore), UNIL-CHUV and Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Davide Malatesta
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Julien F Bally
- Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Aurélien Patoz
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland; Research and Development Department, Volodalen Swiss Sport Lab, Aigle, Switzerland
| |
Collapse
|
18
|
Crotty GF, Ayer SJ, Schwarzschild MA. Designing the First Trials for Parkinson's Prevention. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S381-S393. [PMID: 39302381 PMCID: PMC11491995 DOI: 10.3233/jpd-240164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/20/2024] [Indexed: 09/22/2024]
Abstract
For decades the greatest goal of Parkinson's disease (PD) research has often been distilled to the discovery of treatments that prevent the disease or its progression. However, until recently only the latter has been realistically pursued through randomized clinical trials of candidate disease-modifying therapy (DMT) conducted on individuals after they received traditional clinical diagnosis of PD (i.e., tertiary prevention trials). Now, in light of major advances in our understanding of the prodromal stages of PD, as well as its genetics and biomarkers, the first secondary prevention trials for PD are beginning. In this review, we take stock of DMT trials to date, summarize the breakthroughs that allow the identification of cohorts at high risk of developing a traditional diagnosis of PD, and describe key design elements of secondary prevention trials and how they depend on the prodromal stage being targeted. These elements address whom to enroll, what interventions to test, and how to measure secondary prevention (i.e., slowed progression during the prodromal stages of PD). Although these design strategies, along with the biological definition, subtype classification, and staging of the disease are evolving, all are driven by continued progress in the underlying science and integrated by a broad motivated community of stakeholders. While considerable methodological challenges remain, opportunities to move clinical trials of DMT to earlier points in the disease process than ever before have begun to unfold, and the prospects for PD prevention are nowtangible.
Collapse
Affiliation(s)
- Grace F. Crotty
- Molecular Neurobiology Laboratory, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
- Present address: Department of Neurology, Cork University Hospital, Cork, Ireland
| | - Samuel J. Ayer
- Molecular Neurobiology Laboratory, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Michael A. Schwarzschild
- Molecular Neurobiology Laboratory, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Cen S, Zhang H, Li Y, Gu Z, Yuan Y, Ruan Z, Cai Y, Chhetri JK, Liu S, Mao W, Chan P. Gait Analysis with Wearable Sensors in Isolated REM Sleep Behavior Disorder Associated with Phenoconversion: An Explorative Study. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1027-1037. [PMID: 38848196 PMCID: PMC11307006 DOI: 10.3233/jpd-230397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/09/2024] [Indexed: 06/09/2024]
Abstract
Background Gait disturbance is a vital characteristic of motor manifestation in α- synucleinopathies, especially Parkinson's disease. Subtle gait alterations are present in isolated rapid eye movement sleep behavior disorder (iRBD) patients before phenoconversion; it is yet unclear, if gait analysis may predict phenoconversion. Objective To investigate subtle gait alterations and explore whether gait analysis using wearable sensors is associated with phenoconversion of iRBD to α-synucleinopathies. Methods Thirty-one polysomnography-confirmed iRBD patients and 33 healthy controls (HCs) were enrolled at baseline. All participants walked for a minute while wearing 6 inertial sensors on bilateral wrists, ankles, and the trunk (sternal and lumbar region). Three conditions were tested: (i) normal walking, (ii) fast walking, and (iii) dual-task walking. Results Decreased arm range of motion and increased gait variation (stride length, stride time and stride velocity) discriminate converters from HCs at baseline. After an average of 5.40 years of follow-up, 10 patients converted to neurodegenerative diseases (converters). Cox regression analysis showed higher value of stride length asymmetry under normal walking condition to be associated with an early conversion of iRBD to α- synucleinopathies (adjusted HR 4.468, 95% CI 1.088- 18.349, p = 0.038). Conclusions Stride length asymmetry is associated with progression to α- synucleinopathies in patients with iRBD. Gait analysis with wearable sensors may be useful for screening, monitoring, and risk stratification for disease-modifying therapy trials in patients with iRBD.
Collapse
Affiliation(s)
- Shanshan Cen
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hui Zhang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yuan Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zhuqin Gu
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yuan Yuan
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zheng Ruan
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yanning Cai
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson’s Disease, Parkinson’s Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson’s Disease of Capital Medical University, Beijing, China
- Department of Biobank, Xuanwu Hospital of Capital Medical University, Beijing, China
| | | | - Shuying Liu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wei Mao
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Piu Chan
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
- Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing Key Laboratory on Parkinson’s Disease, Parkinson’s Disease Center for Beijing Institute on Brain Disorders, Clinical and Research Center for Parkinson’s Disease of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| |
Collapse
|
20
|
Lyall DM, Kormilitzin A, Lancaster C, Sousa J, Petermann‐Rocha F, Buckley C, Harshfield EL, Iveson MH, Madan CR, McArdle R, Newby D, Orgeta V, Tang E, Tamburin S, Thakur LS, Lourida I, Llewellyn DJ, Ranson JM. Artificial intelligence for dementia-Applied models and digital health. Alzheimers Dement 2023; 19:5872-5884. [PMID: 37496259 PMCID: PMC10955778 DOI: 10.1002/alz.13391] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/19/2023] [Accepted: 05/26/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION The use of applied modeling in dementia risk prediction, diagnosis, and prognostics will have substantial public health benefits, particularly as "deep phenotyping" cohorts with multi-omics health data become available. METHODS This narrative review synthesizes understanding of applied models and digital health technologies, in terms of dementia risk prediction, diagnostic discrimination, prognosis, and progression. Machine learning approaches show evidence of improved predictive power compared to standard clinical risk scores in predicting dementia, and the potential to decompose large numbers of variables into relatively few critical predictors. RESULTS This review focuses on key areas of emerging promise including: emphasis on easier, more transparent data sharing and cohort access; integration of high-throughput biomarker and electronic health record data into modeling; and progressing beyond the primary prediction of dementia to secondary outcomes, for example, treatment response and physical health. DISCUSSION Such approaches will benefit also from improvements in remote data measurement, whether cognitive (e.g., online), or naturalistic (e.g., watch-based accelerometry).
Collapse
Affiliation(s)
- Donald M. Lyall
- School of Health and WellbeingCollege of Medical and Veterinary Sciences, University of GlasgowGlasgowUK
| | | | | | - Jose Sousa
- Personal Health Data ScienceSANO‐Centre for Computational Personalised MedicineKrakowPoland
- Faculty of MedicineHealth and Life Science, Queen's University BelfastBelfastUK
| | - Fanny Petermann‐Rocha
- School of Health and WellbeingCollege of Medical and Veterinary Sciences, University of GlasgowGlasgowUK
- Centro de Investigación BiomédicaFacultad de Medicina, Universidad Diego PortalesSantiagoChile
| | - Christopher Buckley
- Department of SportExercise and Rehabilitation, Northumbria UniversityNewcastle upon TyneUK
| | - Eric L. Harshfield
- Stroke Research Group, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Matthew H. Iveson
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | | | - Ríona McArdle
- Translational and Clinical Research InstituteFaculty of Medical Sciences, Newcastle UniversityNewcastle upon TyneUK
| | | | | | - Eugene Tang
- Translational and Clinical Research InstituteFaculty of Medical Sciences, Newcastle UniversityNewcastle upon TyneUK
| | - Stefano Tamburin
- Department of NeurosciencesBiomedicine and Movement Sciences, University of VeronaVeronaItaly
| | | | | | | | - David J. Llewellyn
- University of Exeter Medical SchoolExeterUK
- Alan Turing InstituteLondonUK
| | | |
Collapse
|
21
|
Janssen Daalen JM, Oosterhof TH, Bloem BR, Darweesh SKL. Wearables for Parkinson's: Fast-Paced toward Novel Outcome Measures. Mov Disord 2023; 38:1988-1989. [PMID: 37776035 DOI: 10.1002/mds.29608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 10/01/2023] Open
Affiliation(s)
- Jules M Janssen Daalen
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Department of Neurology, Nijmegen, the Netherlands
| | - Thomas H Oosterhof
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Department of Neurology, Nijmegen, the Netherlands
| | - Bastiaan R Bloem
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Department of Neurology, Nijmegen, the Netherlands
| | - Sirwan K L Darweesh
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Department of Neurology, Nijmegen, the Netherlands
| |
Collapse
|
22
|
Jayasinghe U, Hwang F, Harwin WS. Inertial measurement data from loose clothing worn on the lower body during everyday activities. Sci Data 2023; 10:709. [PMID: 37848448 PMCID: PMC10582085 DOI: 10.1038/s41597-023-02567-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/13/2023] [Indexed: 10/19/2023] Open
Abstract
Embedding sensors into clothing is promising as a way for people to wear multiple sensors easily, for applications such as long-term activity monitoring. To our knowledge, this is the first published dataset collected from sensors in loose clothing. 6 Inertial Measurement Units (IMUs) were configured as a 'sensor string' and attached to casual trousers such that there were three sensors on each leg near the waist, thigh, and ankle/lower-shank. Participants also wore an Actigraph accelerometer on their dominant wrist. The dataset consists of 15 participant-days worth of data collected from 5 healthy adults (age range: 28-48 years, 3 males and 2 females). Each participant wore the clothes with sensors for between 1 and 4 days for 5-8 hours per day. Each day, data were collected while participants completed a fixed circuit of activities (with a video ground truth) as well as during free day-to-day activities (with a diary). This dataset can be used to analyse human movements, transitional movements, and postural changes based on a range of features.
Collapse
Affiliation(s)
- Udeni Jayasinghe
- Biomedical Engineering Section, University of Reading, RG6 6DH, Reading, UK.
| | - Faustina Hwang
- Biomedical Engineering Section, University of Reading, RG6 6DH, Reading, UK
| | - William S Harwin
- Biomedical Engineering Section, University of Reading, RG6 6DH, Reading, UK
| |
Collapse
|
23
|
Romijnders R, Salis F, Hansen C, Küderle A, Paraschiv-Ionescu A, Cereatti A, Alcock L, Aminian K, Becker C, Bertuletti S, Bonci T, Brown P, Buckley E, Cantu A, Carsin AE, Caruso M, Caulfield B, Chiari L, D'Ascanio I, Del Din S, Eskofier B, Fernstad SJ, Fröhlich MS, Garcia Aymerich J, Gazit E, Hausdorff JM, Hiden H, Hume E, Keogh A, Kirk C, Kluge F, Koch S, Mazzà C, Megaritis D, Micó-Amigo E, Müller A, Palmerini L, Rochester L, Schwickert L, Scott K, Sharrack B, Singleton D, Soltani A, Ullrich M, Vereijken B, Vogiatzis I, Yarnall A, Schmidt G, Maetzler W. Ecological validity of a deep learning algorithm to detect gait events from real-life walking bouts in mobility-limiting diseases. Front Neurol 2023; 14:1247532. [PMID: 37909030 PMCID: PMC10615212 DOI: 10.3389/fneur.2023.1247532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/18/2023] [Indexed: 11/02/2023] Open
Abstract
Introduction The clinical assessment of mobility, and walking specifically, is still mainly based on functional tests that lack ecological validity. Thanks to inertial measurement units (IMUs), gait analysis is shifting to unsupervised monitoring in naturalistic and unconstrained settings. However, the extraction of clinically relevant gait parameters from IMU data often depends on heuristics-based algorithms that rely on empirically determined thresholds. These were mainly validated on small cohorts in supervised settings. Methods Here, a deep learning (DL) algorithm was developed and validated for gait event detection in a heterogeneous population of different mobility-limiting disease cohorts and a cohort of healthy adults. Participants wore pressure insoles and IMUs on both feet for 2.5 h in their habitual environment. The raw accelerometer and gyroscope data from both feet were used as input to a deep convolutional neural network, while reference timings for gait events were based on the combined IMU and pressure insoles data. Results and discussion The results showed a high-detection performance for initial contacts (ICs) (recall: 98%, precision: 96%) and final contacts (FCs) (recall: 99%, precision: 94%) and a maximum median time error of -0.02 s for ICs and 0.03 s for FCs. Subsequently derived temporal gait parameters were in good agreement with a pressure insoles-based reference with a maximum mean difference of 0.07, -0.07, and <0.01 s for stance, swing, and stride time, respectively. Thus, the DL algorithm is considered successful in detecting gait events in ecologically valid environments across different mobility-limiting diseases.
Collapse
Affiliation(s)
- Robbin Romijnders
- Digital Signal Processing and System Theory, Electrical and Information Engineering, Faculty of Engineering, Kiel University, Kiel, Germany
- Arbeitsgruppe Neurogeriatrie, Department of Neurology, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Francesca Salis
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Clint Hansen
- Arbeitsgruppe Neurogeriatrie, Department of Neurology, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Arne Küderle
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anisoara Paraschiv-Ionescu
- Laboratory of Movement Analysis and Measurement, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Andrea Cereatti
- Department of Electronics and Telecommunications, Polytechnic of Turin, Turin, Italy
| | - Lisa Alcock
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kamiar Aminian
- Laboratory of Movement Analysis and Measurement, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Clemens Becker
- Gesellschaft für Medizinische Forschung, Robert-Bosch Foundation GmbH, Stuttgart, Germany
| | - Stefano Bertuletti
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Tecla Bonci
- INSIGNEO Institute for In Silico Medicine, The University of Sheffield, Sheffield, United Kingdom
- Department of Mechanical Engineering, The University of Sheffield, Sheffield, United Kingdom
| | - Philip Brown
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Ellen Buckley
- INSIGNEO Institute for In Silico Medicine, The University of Sheffield, Sheffield, United Kingdom
- Department of Mechanical Engineering, The University of Sheffield, Sheffield, United Kingdom
| | - Alma Cantu
- School of Computing, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anne-Elie Carsin
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Faculty of Health and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Marco Caruso
- Department of Electronics and Telecommunications, Polytechnic of Turin, Turin, Italy
| | - Brian Caulfield
- Insight Centre for Data Analytics, University College Dublin, Dublin, Ireland
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| | - Lorenzo Chiari
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi”, University of Bologna, Bologna, Italy
- Health Sciences and Technologies—Interdepartmental Center for Industrial Research (CIRISDV), University of Bologna, Bologna, Italy
| | - Ilaria D'Ascanio
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi”, University of Bologna, Bologna, Italy
| | - Silvia Del Din
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Björn Eskofier
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | - Judith Garcia Aymerich
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Faculty of Health and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Eran Gazit
- Center for the Study of Movement, Cognition and Mobility, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Jeffrey M. Hausdorff
- Center for the Study of Movement, Cognition and Mobility, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Department of Physical Therapy, Sackler Faculty of Medicine & Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Hugo Hiden
- School of Computing, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Emily Hume
- Department of Sport, Exercise and Rehabilitation, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Alison Keogh
- Insight Centre for Data Analytics, University College Dublin, Dublin, Ireland
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| | - Cameron Kirk
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Felix Kluge
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Novartis Institute of Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Sarah Koch
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Faculty of Health and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Claudia Mazzà
- INSIGNEO Institute for In Silico Medicine, The University of Sheffield, Sheffield, United Kingdom
- Department of Mechanical Engineering, The University of Sheffield, Sheffield, United Kingdom
| | - Dimitrios Megaritis
- Department of Sport, Exercise and Rehabilitation, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Encarna Micó-Amigo
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Arne Müller
- Novartis Institute of Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Luca Palmerini
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi”, University of Bologna, Bologna, Italy
- Health Sciences and Technologies—Interdepartmental Center for Industrial Research (CIRISDV), University of Bologna, Bologna, Italy
| | - Lynn Rochester
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Lars Schwickert
- Gesellschaft für Medizinische Forschung, Robert-Bosch Foundation GmbH, Stuttgart, Germany
| | - Kirsty Scott
- INSIGNEO Institute for In Silico Medicine, The University of Sheffield, Sheffield, United Kingdom
- Department of Mechanical Engineering, The University of Sheffield, Sheffield, United Kingdom
| | - Basil Sharrack
- Department of Neuroscience and Sheffield NIHR Translational Neuroscience BRC, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - David Singleton
- Insight Centre for Data Analytics, University College Dublin, Dublin, Ireland
- School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| | - Abolfazl Soltani
- Laboratory of Movement Analysis and Measurement, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Digital Health Department, CSEM SA, Neuchâtel, Switzerland
| | - Martin Ullrich
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Beatrix Vereijken
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ioannis Vogiatzis
- Department of Sport, Exercise and Rehabilitation, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Alison Yarnall
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Mechanical Engineering, The University of Sheffield, Sheffield, United Kingdom
| | - Gerhard Schmidt
- Digital Signal Processing and System Theory, Electrical and Information Engineering, Faculty of Engineering, Kiel University, Kiel, Germany
| | - Walter Maetzler
- Arbeitsgruppe Neurogeriatrie, Department of Neurology, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
24
|
Sotirakis C, Su Z, Brzezicki MA, Conway N, Tarassenko L, FitzGerald JJ, Antoniades CA. Identification of motor progression in Parkinson's disease using wearable sensors and machine learning. NPJ Parkinsons Dis 2023; 9:142. [PMID: 37805655 PMCID: PMC10560243 DOI: 10.1038/s41531-023-00581-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 09/20/2023] [Indexed: 10/09/2023] Open
Abstract
Wearable devices offer the potential to track motor symptoms in neurological disorders. Kinematic data used together with machine learning algorithms can accurately identify people living with movement disorders and the severity of their motor symptoms. In this study we aimed to establish whether a combination of wearable sensor data and machine learning algorithms with automatic feature selection can estimate the clinical rating scale and whether it is possible to monitor the motor symptom progression longitudinally, for people with Parkinson's Disease. Seventy-four patients visited the lab seven times at 3-month intervals. Their walking (2-minutes) and postural sway (30-seconds,eyes-closed) were recorded using six Inertial Measurement Unit sensors. Simple linear regression and Random Forest algorithms were utilised together with different routines of automatic feature selection or factorisation, resulting in seven different machine learning algorithms to estimate the clinical rating scale (Movement Disorder Society- Unified Parkinson's Disease Rating Scale part III; MDS-UPDRS-III). Twenty-nine features were found to significantly progress with time at group level. The Random Forest model revealed the most accurate estimation of the MDS-UPDRS-III among the seven models. The model estimations detected a statistically significant progression of the motor symptoms within 15 months when compared to the first visit, whereas the MDS-UPDRS-III did not capture any change. Wearable sensors and machine learning can track the motor symptom progression in people with PD better than the conventionally used clinical rating scales. The methods described in this study can be utilised complimentary to the clinical rating scales to improve the diagnostic and prognostic accuracy.
Collapse
Affiliation(s)
- Charalampos Sotirakis
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Zi Su
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Maksymilian A Brzezicki
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Niall Conway
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Lionel Tarassenko
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - James J FitzGerald
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Chrystalina A Antoniades
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| |
Collapse
|
25
|
Canonico M, Desimoni F, Ferrero A, Grassi PA, Irwin C, Campani D, Dal Molin A, Panella M, Magistrelli L. Gait Monitoring and Analysis: A Mathematical Approach. SENSORS (BASEL, SWITZERLAND) 2023; 23:7743. [PMID: 37765801 PMCID: PMC10536663 DOI: 10.3390/s23187743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/29/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023]
Abstract
Gait abnormalities are common in the elderly and individuals diagnosed with Parkinson's, often leading to reduced mobility and increased fall risk. Monitoring and assessing gait patterns in these populations play a crucial role in understanding disease progression, early detection of motor impairments, and developing personalized rehabilitation strategies. In particular, by identifying gait irregularities at an early stage, healthcare professionals can implement timely interventions and personalized therapeutic approaches, potentially delaying the onset of severe motor symptoms and improving overall patient outcomes. In this paper, we studied older adults affected by chronic diseases and/or Parkinson's disease by monitoring their gait due to wearable devices that can accurately detect a person's movements. In our study, about 50 people were involved in the trial (20 with Parkinson's disease and 30 people with chronic diseases) who have worn our device for at least 6 months. During the experimentation, each device collected 25 samples from the accelerometer sensor for each second. By analyzing those data, we propose a metric for the "gait quality" based on the measure of entropy obtained by applying the Fourier transform.
Collapse
Affiliation(s)
- Massimo Canonico
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 15121 Alessandria, Italy; (F.D.); (A.F.); (P.A.G.); (C.I.)
| | - Francesco Desimoni
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 15121 Alessandria, Italy; (F.D.); (A.F.); (P.A.G.); (C.I.)
| | - Alberto Ferrero
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 15121 Alessandria, Italy; (F.D.); (A.F.); (P.A.G.); (C.I.)
| | - Pietro Antonio Grassi
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 15121 Alessandria, Italy; (F.D.); (A.F.); (P.A.G.); (C.I.)
| | - Christopher Irwin
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 15121 Alessandria, Italy; (F.D.); (A.F.); (P.A.G.); (C.I.)
| | - Daiana Campani
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.C.); (A.D.M.); (M.P.); (L.M.)
| | - Alberto Dal Molin
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.C.); (A.D.M.); (M.P.); (L.M.)
| | - Massimiliano Panella
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.C.); (A.D.M.); (M.P.); (L.M.)
| | - Luca Magistrelli
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.C.); (A.D.M.); (M.P.); (L.M.)
| |
Collapse
|
26
|
Tam W, Alajlani M, Abd-Alrazaq A. An Exploration of Wearable Device Features Used in UK Hospital Parkinson Disease Care: Scoping Review. J Med Internet Res 2023; 25:e42950. [PMID: 37594791 PMCID: PMC10474516 DOI: 10.2196/42950] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 03/13/2023] [Accepted: 04/14/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND The prevalence of Parkinson disease (PD) is becoming an increasing concern owing to the aging population in the United Kingdom. Wearable devices have the potential to improve the clinical care of patients with PD while reducing health care costs. Consequently, exploring the features of these wearable devices is important to identify the limitations and further areas of investigation of how wearable devices are currently used in clinical care in the United Kingdom. OBJECTIVE In this scoping review, we aimed to explore the features of wearable devices used for PD in hospitals in the United Kingdom. METHODS A scoping review of the current research was undertaken and reported according to the PRISMA-ScR (Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews) guidelines. The literature search was undertaken on June 6, 2022, and publications were obtained from MEDLINE or PubMed, Embase, and the Cochrane Library. Eligible publications were initially screened by their titles and abstracts. Publications that passed the initial screening underwent a full review. The study characteristics were extracted from the final publications, and the evidence was synthesized using a narrative approach. Any queries were reviewed by the first and second authors. RESULTS Of the 4543 publications identified, 39 (0.86%) publications underwent a full review, and 20 (0.44%) publications were included in the scoping review. Most studies (11/20, 55%) were conducted at the Newcastle upon Tyne Hospitals NHS Foundation Trust, with sample sizes ranging from 10 to 418. Most study participants were male individuals with a mean age ranging from 57.7 to 78.0 years. The AX3 was the most popular device brand used, and it was commercially manufactured by Axivity. Common wearable device types included body-worn sensors, inertial measurement units, and smartwatches that used accelerometers and gyroscopes to measure the clinical features of PD. Most wearable device primary measures involved the measured gait, bradykinesia, and dyskinesia. The most common wearable device placements were the lumbar region, head, and wrist. Furthermore, 65% (13/20) of the studies used artificial intelligence or machine learning to support PD data analysis. CONCLUSIONS This study demonstrated that wearable devices could help provide a more detailed analysis of PD symptoms during the assessment phase and personalize treatment. Using machine learning, wearable devices could differentiate PD from other neurodegenerative diseases. The identified evidence gaps include the lack of analysis of wearable device cybersecurity and data management. The lack of cost-effectiveness analysis and large-scale participation in studies resulted in uncertainty regarding the feasibility of the widespread use of wearable devices. The uncertainty around the identified research gaps was further exacerbated by the lack of medical regulation of wearable devices for PD, particularly in the United Kingdom where regulations were changing due to the political landscape.
Collapse
Affiliation(s)
- William Tam
- Insitute of Digital Healthcare, Warwick Manufacturing Group, University of Warwick, Coventry, United Kingdom
| | - Mohannad Alajlani
- Insitute of Digital Healthcare, Warwick Manufacturing Group, University of Warwick, Coventry, United Kingdom
| | | |
Collapse
|
27
|
Schalkamp AK, Peall KJ, Harrison NA, Sandor C. Wearable movement-tracking data identify Parkinson's disease years before clinical diagnosis. Nat Med 2023; 29:2048-2056. [PMID: 37400639 DOI: 10.1038/s41591-023-02440-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/05/2023] [Indexed: 07/05/2023]
Abstract
Parkinson's disease is a progressive neurodegenerative movement disorder with a long latent phase and currently no disease-modifying treatments. Reliable predictive biomarkers that could transform efforts to develop neuroprotective treatments remain to be identified. Using UK Biobank, we investigated the predictive value of accelerometry in identifying prodromal Parkinson's disease in the general population and compared this digital biomarker with models based on genetics, lifestyle, blood biochemistry or prodromal symptoms data. Machine learning models trained using accelerometry data achieved better test performance in distinguishing both clinically diagnosed Parkinson's disease (n = 153) (area under precision recall curve (AUPRC) 0.14 ± 0.04) and prodromal Parkinson's disease (n = 113) up to 7 years pre-diagnosis (AUPRC 0.07 ± 0.03) from the general population (n = 33,009) compared with all other modalities tested (genetics: AUPRC = 0.01 ± 0.00, P = 2.2 × 10-3; lifestyle: AUPRC = 0.03 ± 0.04, P = 2.5 × 10-3; blood biochemistry: AUPRC = 0.01 ± 0.00, P = 4.1 × 10-3; prodromal signs: AUPRC = 0.01 ± 0.00, P = 3.6 × 10-3). Accelerometry is a potentially important, low-cost screening tool for determining people at risk of developing Parkinson's disease and identifying participants for clinical trials of neuroprotective treatments.
Collapse
Affiliation(s)
- Ann-Kathrin Schalkamp
- Division of Psychological Medicine and Clinical Neuroscience, UK Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Kathryn J Peall
- Division of Psychological Medicine and Clinical Neurosciences, Neuroscience and Mental Health Innovation Institute, Cardiff, UK
| | - Neil A Harrison
- Division of Psychological Medicine and Clinical Neurosciences, Neuroscience and Mental Health Innovation Institute, Cardiff, UK
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff, UK
| | - Cynthia Sandor
- Division of Psychological Medicine and Clinical Neuroscience, UK Dementia Research Institute, Cardiff University, Cardiff, UK.
| |
Collapse
|
28
|
Payne T, Appleby M, Buckley E, van Gelder LM, Mullish BH, Sassani M, Dunning MJ, Hernandez D, Scholz S, McNeil A, Libri V, Moll S, Marchesi JR, Taylor R, Su L, Mazzà C, Jenkins TM, Foltynie T, Bandmann O. A Double-Blind, Randomized, Placebo-Controlled Trial of Ursodeoxycholic Acid (UDCA) in Parkinson's Disease. Mov Disord 2023; 38:1493-1502. [PMID: 37246815 PMCID: PMC10527073 DOI: 10.1002/mds.29450] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Rescue of mitochondrial function is a promising neuroprotective strategy for Parkinson's disease (PD). Ursodeoxycholic acid (UDCA) has shown considerable promise as a mitochondrial rescue agent across a range of preclinical in vitro and in vivo models of PD. OBJECTIVES To investigate the safety and tolerability of high-dose UDCA in PD and determine midbrain target engagement. METHODS The UP (UDCA in PD) study was a phase II, randomized, double-blind, placebo-controlled trial of UDCA (30 mg/kg daily, 2:1 randomization UDCA vs. placebo) in 30 participants with PD for 48 weeks. The primary outcome was safety and tolerability. Secondary outcomes included 31-phosphorus magnetic resonance spectroscopy (31 P-MRS) to explore target engagement of UDCA in PD midbrain and assessment of motor progression, applying both the Movement Disorder Society Unified Parkinson's Disease Rating Scale Part III (MDS-UPDRS-III) and objective, motion sensor-based quantification of gait impairment. RESULTS UDCA was safe and well tolerated, and only mild transient gastrointestinal adverse events were more frequent in the UDCA treatment group. Midbrain 31 P-MRS demonstrated an increase in both Gibbs free energy and inorganic phosphate levels in the UDCA treatment group compared to placebo, reflecting improved ATP hydrolysis. Sensor-based gait analysis indicated a possible improvement of cadence (steps per minute) and other gait parameters in the UDCA group compared to placebo. In contrast, subjective assessment applying the MDS-UPDRS-III failed to detect a difference between treatment groups. CONCLUSIONS High-dose UDCA is safe and well tolerated in early PD. Larger trials are needed to further evaluate the disease-modifying effect of UDCA in PD. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Thomas Payne
- Sheffield Institute for Translational Neuroscience,
University of Sheffield, Sheffield, S10 2HQ, United Kingdom
| | - Matthew Appleby
- NIHR UCLH Clinical Research Facility – Leonard
Wolfson Experimental Neurology Centre, National Hospital for Neurology &
Neurosurgery, London, WC1N 3BG, United Kingdom
- Department of Clinical and Movement Neurosciences,
Institute of Neurology, University College London, London, WC1N 3BG, United
Kingdom
| | - Ellen Buckley
- Department of Mechanical Engineering and Insigneo Institute
for In Silico Medicine, The University of Sheffield, Sheffield, S1 3JD, United
Kingdom
| | - Linda M.A. van Gelder
- Department of Mechanical Engineering and Insigneo Institute
for In Silico Medicine, The University of Sheffield, Sheffield, S1 3JD, United
Kingdom
| | - Benjamin H. Mullish
- Division of Digestive Diseases, Department of Metabolism,
Digestion and Reproduction, St Mary’s Hospital Campus, Imperial College
London, London, W2 1NY, United Kingdom
| | - Matilde Sassani
- Sheffield Institute for Translational Neuroscience,
University of Sheffield, Sheffield, S10 2HQ, United Kingdom
| | - Mark J. Dunning
- Sheffield Institute for Translational Neuroscience,
University of Sheffield, Sheffield, S10 2HQ, United Kingdom
- The Bioinformatics Core, Sheffield Institute of
Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, United
Kingdom
| | - Dena Hernandez
- Molecular Genetics Section, Laboratory of Neurogenetics,
NIA, NIH, Bethesda, Maryland, MD 20814, USA
| | - Sonja Scholz
- Neurodegenerative Diseases Research Unit, Laboratory of
Neurogenetics, National Institute of Neurological Disorders and Stroke, National
Institutes of Health, Bethesda, Maryland, MD 20814, USA
- Department of Neurology, Johns Hopkins University Medical
Center, Baltimore, Maryland, MD 21287, USA
| | - Alisdair McNeil
- Sheffield Institute for Translational Neuroscience,
University of Sheffield, Sheffield, S10 2HQ, United Kingdom
| | - Vincenzo Libri
- NIHR UCLH Clinical Research Facility – Leonard
Wolfson Experimental Neurology Centre, National Hospital for Neurology &
Neurosurgery, London, WC1N 3BG, United Kingdom
| | - Sarah Moll
- NIHR Sheffield Biomedical Research Centre, Royal
Hallamshire Hospital, Sheffield, S10 2JF United Kingdom
| | - Julian R. Marchesi
- Division of Digestive Diseases, Department of Metabolism,
Digestion and Reproduction, St Mary’s Hospital Campus, Imperial College
London, London, W2 1NY, United Kingdom
| | - Rosie Taylor
- Statistical Services Unit, The University of Sheffield,
Sheffield, S3 7RH, United Kingdom
| | - Li Su
- Sheffield Institute for Translational Neuroscience,
University of Sheffield, Sheffield, S10 2HQ, United Kingdom
- Department of Psychiatry, University of Cambridge, CB2
0SP United Kingdom
| | - Claudia Mazzà
- Department of Mechanical Engineering and Insigneo Institute
for In Silico Medicine, The University of Sheffield, Sheffield, S1 3JD, United
Kingdom
| | - Thomas M. Jenkins
- Sheffield Institute for Translational Neuroscience,
University of Sheffield, Sheffield, S10 2HQ, United Kingdom
- Royal Perth Hospital, Victoria Square, Perth, WA 6000,
Australia
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences,
Institute of Neurology, University College London, London, WC1N 3BG, United
Kingdom
| | - Oliver Bandmann
- Sheffield Institute for Translational Neuroscience,
University of Sheffield, Sheffield, S10 2HQ, United Kingdom
| |
Collapse
|
29
|
Manto M, Serrao M, Filippo Castiglia S, Timmann D, Tzvi-Minker E, Pan MK, Kuo SH, Ugawa Y. Neurophysiology of cerebellar ataxias and gait disorders. Clin Neurophysiol Pract 2023; 8:143-160. [PMID: 37593693 PMCID: PMC10429746 DOI: 10.1016/j.cnp.2023.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/19/2023] [Accepted: 07/11/2023] [Indexed: 08/19/2023] Open
Abstract
There are numerous forms of cerebellar disorders from sporadic to genetic diseases. The aim of this chapter is to provide an overview of the advances and emerging techniques during these last 2 decades in the neurophysiological tests useful in cerebellar patients for clinical and research purposes. Clinically, patients exhibit various combinations of a vestibulocerebellar syndrome, a cerebellar cognitive affective syndrome and a cerebellar motor syndrome which will be discussed throughout this chapter. Cerebellar patients show abnormal Bereitschaftpotentials (BPs) and mismatch negativity. Cerebellar EEG is now being applied in cerebellar disorders to unravel impaired electrophysiological patterns associated within disorders of the cerebellar cortex. Eyeblink conditioning is significantly impaired in cerebellar disorders: the ability to acquire conditioned eyeblink responses is reduced in hereditary ataxias, in cerebellar stroke and after tumor surgery of the cerebellum. Furthermore, impaired eyeblink conditioning is an early marker of cerebellar degenerative disease. General rules of motor control suggest that optimal strategies are needed to execute voluntary movements in the complex environment of daily life. A high degree of adaptability is required for learning procedures underlying motor control as sensorimotor adaptation is essential to perform accurate goal-directed movements. Cerebellar patients show impairments during online visuomotor adaptation tasks. Cerebellum-motor cortex inhibition (CBI) is a neurophysiological biomarker showing an inverse association between cerebellothalamocortical tract integrity and ataxia severity. Ataxic gait is characterized by increased step width, reduced ankle joint range of motion, increased gait variability, lack of intra-limb inter-joint and inter-segmental coordination, impaired foot ground placement and loss of trunk control. Taken together, these techniques provide a neurophysiological framework for a better appraisal of cerebellar disorders.
Collapse
Affiliation(s)
- Mario Manto
- Service des Neurosciences, Université de Mons, Mons, Belgium
- Service de Neurologie, CHU-Charleroi, Charleroi, Belgium
| | - Mariano Serrao
- Department of Medical and Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, Corso della Repubblica 79 04100, Latina, Italy
- Gait Analysis LAB Policlinico Italia, Via Del Campidano 6 00162, Rome, Italy
| | - Stefano Filippo Castiglia
- Department of Medical and Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, Corso della Repubblica 79 04100, Latina, Italy
- Gait Analysis LAB Policlinico Italia, Via Del Campidano 6 00162, Rome, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, via Bassi, 21, 27100 Pavia, Italy
| | - Dagmar Timmann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Elinor Tzvi-Minker
- Department of Neurology, University of Leipzig, Liebigstraße 20, 04103 Leipzig, Germany
- Syte Institute, Hamburg, Germany
| | - Ming-Kai Pan
- Cerebellar Research Center, National Taiwan University Hospital, Yun-Lin Branch, Yun-Lin 64041, Taiwan
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei 10051, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei 10002, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei City 11529, Taiwan
- Initiative for Columbia Ataxia and Tremor, Columbia University Irving Medical Center, New York, NY, USA
| | - Sheng-Han Kuo
- Institute of Biomedical Sciences, Academia Sinica, Taipei City 11529, Taiwan
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Yoshikazu Ugawa
- Department of Human Neurophysiology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
30
|
Sun Y, Cheng Y, You Y, Wang Y, Zhu Z, Yu Y, Han J, Wu J, Yu N. A novel plantar pressure analysis method to signify gait dynamics in Parkinson's disease. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:13474-13490. [PMID: 37679098 DOI: 10.3934/mbe.2023601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Plantar pressure can signify the gait performance of patients with Parkinson's disease (PD). This study proposed a plantar pressure analysis method with the dynamics feature of the sub-regions plantar pressure signals. Specifically, each side's plantar pressure signals were divided into five sub-regions. Moreover, a dynamics feature extractor (DFE) was designed to extract features of the sub-regions signals. The radial basis function neural network (RBFNN) was used to learn and store gait dynamics. And a classification mechanism based on the output error in RBFNN was proposed. The classification accuracy of the proposed method achieved 100.00% in PD diagnosis and 95.89% in severity assessment on the online dataset, and 96.00% in severity assessment on our dataset. The experimental results suggested that the proposed method had the capability to signify the gait dynamics of PD patients.
Collapse
Affiliation(s)
- Yubo Sun
- College of Artificial Intelligence, Nankai University, Tianjin 300350, China
| | - Yuanyuan Cheng
- Department of Rehabilitation Medicine, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Yugen You
- College of Artificial Intelligence, Nankai University, Tianjin 300350, China
| | - Yue Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300070, China
| | - Zhizhong Zhu
- Department of Rehabilitation Medicine, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Yang Yu
- Department of Rehabilitation Medicine, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Jianda Han
- College of Artificial Intelligence, Nankai University, Tianjin 300350, China
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Nankai University, Tianjin 300350, China
- Tianjin Key Laboratory of Intelligent Robotics, Nankai University, Tianjin 300350, China
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Shenzhen 518083, China
| | - Jialing Wu
- Department of Rehabilitation Medicine, Tianjin Huanhu Hospital, Tianjin 300350, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Ningbo Yu
- College of Artificial Intelligence, Nankai University, Tianjin 300350, China
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Nankai University, Tianjin 300350, China
- Tianjin Key Laboratory of Intelligent Robotics, Nankai University, Tianjin 300350, China
- Institute of Intelligence Technology and Robotic Systems, Shenzhen Research Institute of Nankai University, Shenzhen 518083, China
| |
Collapse
|
31
|
Paulauskaite-Taraseviciene A, Siaulys J, Sutiene K, Petravicius T, Navickas S, Oliandra M, Rapalis A, Balciunas J. Geriatric Care Management System Powered by the IoT and Computer Vision Techniques. Healthcare (Basel) 2023; 11:1152. [PMID: 37107987 PMCID: PMC10138364 DOI: 10.3390/healthcare11081152] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
The digitalisation of geriatric care refers to the use of emerging technologies to manage and provide person-centered care to the elderly by collecting patients' data electronically and using them to streamline the care process, which improves the overall quality, accuracy, and efficiency of healthcare. In many countries, healthcare providers still rely on the manual measurement of bioparameters, inconsistent monitoring, and paper-based care plans to manage and deliver care to elderly patients. This can lead to a number of problems, including incomplete and inaccurate record-keeping, errors, and delays in identifying and resolving health problems. The purpose of this study is to develop a geriatric care management system that combines signals from various wearable sensors, noncontact measurement devices, and image recognition techniques to monitor and detect changes in the health status of a person. The system relies on deep learning algorithms and the Internet of Things (IoT) to identify the patient and their six most pertinent poses. In addition, the algorithm has been developed to monitor changes in the patient's position over a longer period of time, which could be important for detecting health problems in a timely manner and taking appropriate measures. Finally, based on expert knowledge and a priori rules integrated in a decision tree-based model, the automated final decision on the status of nursing care plan is generated to support nursing staff.
Collapse
Affiliation(s)
| | - Julius Siaulys
- Faculty of Informatics, Kaunas University of Technology, Studentu 50, 51368 Kaunas, Lithuania
| | - Kristina Sutiene
- Department of Mathematical Modeling, Kaunas University of Technology, Studentu 50, 51368 Kaunas, Lithuania
| | - Titas Petravicius
- Faculty of Informatics, Kaunas University of Technology, Studentu 50, 51368 Kaunas, Lithuania
| | - Skirmantas Navickas
- Faculty of Informatics, Kaunas University of Technology, Studentu 50, 51368 Kaunas, Lithuania
| | - Marius Oliandra
- Faculty of Informatics, Kaunas University of Technology, Studentu 50, 51368 Kaunas, Lithuania
| | - Andrius Rapalis
- Biomedical Engineering Institute, Kaunas University of Technology, K. Barsausko 59, 51423 Kaunas, Lithuania
- Faculty of Electrical and Electronics Engineering, Kaunas University of Technology, Studentu 48, 51367 Kaunas, Lithuania
| | - Justinas Balciunas
- Faculty of Medicine, Vilnius University, Universiteto 3, 01513 Vilnius, Lithuania
| |
Collapse
|
32
|
Debelle H, Packer E, Beales E, Bailey HGB, Mc Ardle R, Brown P, Hunter H, Ciravegna F, Ireson N, Evers J, Niessen M, Shi JQ, Yarnall AJ, Rochester L, Alcock L, Del Din S. Feasibility and usability of a digital health technology system to monitor mobility and assess medication adherence in mild-to-moderate Parkinson's disease. Front Neurol 2023; 14:1111260. [PMID: 37006505 PMCID: PMC10050691 DOI: 10.3389/fneur.2023.1111260] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/20/2023] [Indexed: 03/17/2023] Open
Abstract
IntroductionParkinson's disease (PD) is a neurodegenerative disorder which requires complex medication regimens to mitigate motor symptoms. The use of digital health technology systems (DHTSs) to collect mobility and medication data provides an opportunity to objectively quantify the effect of medication on motor performance during day-to-day activities. This insight could inform clinical decision-making, personalise care, and aid self-management. This study investigates the feasibility and usability of a multi-component DHTS to remotely assess self-reported medication adherence and monitor mobility in people with Parkinson's (PwP).MethodsThirty participants with PD [Hoehn and Yahr stage I (n = 1) and II (n = 29)] were recruited for this cross-sectional study. Participants were required to wear, and where appropriate, interact with a DHTS (smartwatch, inertial measurement unit, and smartphone) for seven consecutive days to assess medication adherence and monitor digital mobility outcomes and contextual factors. Participants reported their daily motor complications [motor fluctuations and dyskinesias (i.e., involuntary movements)] in a diary. Following the monitoring period, participants completed a questionnaire to gauge the usability of the DHTS. Feasibility was assessed through the percentage of data collected, and usability through analysis of qualitative questionnaire feedback.ResultsAdherence to each device exceeded 70% and ranged from 73 to 97%. Overall, the DHTS was well tolerated with 17/30 participants giving a score > 75% [average score for these participants = 89%, from 0 (worst) to 100 (best)] for its usability. Usability of the DHTS was significantly associated with age (ρ = −0.560, BCa 95% CI [−0.791, −0.207]). This study identified means to improve usability of the DHTS by addressing technical and design issues of the smartwatch. Feasibility, usability and acceptability were identified as key themes from PwP qualitative feedback on the DHTS.ConclusionThis study highlighted the feasibility and usability of our integrated DHTS to remotely assess medication adherence and monitor mobility in people with mild-to-moderate Parkinson's disease. Further work is necessary to determine whether this DHTS can be implemented for clinical decision-making to optimise management of PwP.
Collapse
Affiliation(s)
- Héloïse Debelle
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Emma Packer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Esther Beales
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Harry G. B. Bailey
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ríona Mc Ardle
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- National Institute for Health and Care Research (NIHR), Newcastle Biomedical Research Centre (BRC), Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Philip Brown
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Heather Hunter
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Fabio Ciravegna
- Department of Computer Science and INSIGNEO Institute for in silico Medicine, The University of Sheffield, Sheffield, United Kingdom
- Dipartimento di Informatica, Università di Torino, Turin, Italy
| | - Neil Ireson
- Department of Computer Science and INSIGNEO Institute for in silico Medicine, The University of Sheffield, Sheffield, United Kingdom
| | | | | | - Jian Qing Shi
- Department of Statistics and Data Science, Southern University of Science and Technology, Shenzhen, China
| | - Alison J. Yarnall
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- National Institute for Health and Care Research (NIHR), Newcastle Biomedical Research Centre (BRC), Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Lynn Rochester
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- National Institute for Health and Care Research (NIHR), Newcastle Biomedical Research Centre (BRC), Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Lisa Alcock
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- National Institute for Health and Care Research (NIHR), Newcastle Biomedical Research Centre (BRC), Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Silvia Del Din
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- National Institute for Health and Care Research (NIHR), Newcastle Biomedical Research Centre (BRC), Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- *Correspondence: Silvia Del Din
| |
Collapse
|
33
|
Oyama G, Burq M, Hatano T, Marks WJ, Kapur R, Fernandez J, Fujikawa K, Furusawa Y, Nakatome K, Rainaldi E, Chen C, Ho KC, Ogawa T, Kamo H, Oji Y, Takeshige-Amano H, Taniguchi D, Nakamura R, Sasaki F, Ueno S, Shiina K, Hattori A, Nishikawa N, Ishiguro M, Saiki S, Hayashi A, Motohashi M, Hattori N. Analytical and clinical validity of wearable, multi-sensor technology for assessment of motor function in patients with Parkinson's disease in Japan. Sci Rep 2023; 13:3600. [PMID: 36918552 PMCID: PMC10015076 DOI: 10.1038/s41598-023-29382-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/03/2023] [Indexed: 03/16/2023] Open
Abstract
Continuous, objective monitoring of motor signs and symptoms may help improve tracking of disease progression and treatment response in Parkinson's disease (PD). This study assessed the analytical and clinical validity of multi-sensor smartwatch measurements in hospitalized and home-based settings (96 patients with PD; mean wear time 19 h/day) using a twice-daily virtual motor examination (VME) at times representing medication OFF/ON states. Digital measurement performance was better during inpatient clinical assessments for composite V-scores than single-sensor-derived features for bradykinesia (Spearman |r|= 0.63, reliability = 0.72), tremor (|r|= 0.41, reliability = 0.65), and overall motor features (|r|= 0.70, reliability = 0.67). Composite levodopa effect sizes during hospitalization were 0.51-1.44 for clinical assessments and 0.56-1.37 for VMEs. Reliability of digital measurements during home-based VMEs was 0.62-0.80 for scores derived from weekly averages and 0.24-0.66 for daily measurements. These results show that unsupervised digital measurements of motor features with wrist-worn sensors are sensitive to medication state and are reliable in naturalistic settings.Trial Registration: Japan Pharmaceutical Information Center Clinical Trials Information (JAPIC-CTI): JapicCTI-194825; Registered June 25, 2019.
Collapse
Affiliation(s)
- Genko Oyama
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| | - Maximilien Burq
- Verily Life Sciences, 269 East Grand Avenue, South San Francisco, CA, USA
| | - Taku Hatano
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - William J Marks
- Verily Life Sciences, 269 East Grand Avenue, South San Francisco, CA, USA
| | - Ritu Kapur
- Verily Life Sciences, 269 East Grand Avenue, South San Francisco, CA, USA
| | - Jovelle Fernandez
- Takeda Pharmaceutical Company Limited, 2 Chome-1-1 Nihonbashihoncho, Chuo-Ku, Tokyo, 103-0023, Japan
| | - Keita Fujikawa
- Takeda Pharmaceutical Company Limited, 2 Chome-1-1 Nihonbashihoncho, Chuo-Ku, Tokyo, 103-0023, Japan
| | - Yoshihiko Furusawa
- Takeda Pharmaceutical Company Limited, 2 Chome-1-1 Nihonbashihoncho, Chuo-Ku, Tokyo, 103-0023, Japan
| | - Keisuke Nakatome
- Takeda Pharmaceutical Company Limited, 2 Chome-1-1 Nihonbashihoncho, Chuo-Ku, Tokyo, 103-0023, Japan
| | - Erin Rainaldi
- Verily Life Sciences, 269 East Grand Avenue, South San Francisco, CA, USA
| | - Chen Chen
- Verily Life Sciences, 269 East Grand Avenue, South San Francisco, CA, USA
| | - King Chung Ho
- Verily Life Sciences, 269 East Grand Avenue, South San Francisco, CA, USA
| | - Takashi Ogawa
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Hikaru Kamo
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Yutaka Oji
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Haruka Takeshige-Amano
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Daisuke Taniguchi
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Ryota Nakamura
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Fuyuko Sasaki
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Shinichi Ueno
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Kenta Shiina
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Anri Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Noriko Nishikawa
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Mayu Ishiguro
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Shinji Saiki
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Ayako Hayashi
- Takeda Pharmaceutical Company Limited, 2 Chome-1-1 Nihonbashihoncho, Chuo-Ku, Tokyo, 103-0023, Japan
| | - Masatoshi Motohashi
- Takeda Pharmaceutical Company Limited, 2 Chome-1-1 Nihonbashihoncho, Chuo-Ku, Tokyo, 103-0023, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, 2-1-1, Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| |
Collapse
|
34
|
Chahine LM, Simuni T. Role of novel endpoints and evaluations of response in Parkinson disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:325-345. [PMID: 36803820 DOI: 10.1016/b978-0-323-85555-6.00010-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
With progress in our understanding of Parkinson disease (PD) and other neurodegenerative disorders, from clinical features to imaging, genetic, and molecular characterization comes the opportunity to refine and revise how we measure these diseases and what outcome measures are used as endpoints in clinical trials. While several rater-, patient-, and milestone-based outcomes for PD exist that may serve as clinical trial endpoints, there remains an unmet need for endpoints that are clinically meaningful, patient centric while also being more objective and quantitative, less susceptible to effects of symptomatic therapy (for disease-modification trials), and that can be measured over a short period and yet accurately represent longer-term outcomes. Several novel outcomes that may be used as endpoints in PD clinical trials are in development, including digital measures of signs and symptoms, as well a growing array of imaging and biospecimen biomarkers. This chapter provides an overview of the state of PD outcome measures as of 2022, including considerations for selection of clinical trial endpoints in PD, advantages and limitations of existing measures, and emerging potential novel endpoints.
Collapse
Affiliation(s)
- Lana M Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.
| |
Collapse
|
35
|
Su ZH, Patel S, Gavine B, Buchanan T, Bogdanovic M, Sarangmat N, Green AL, Bloem BR, FitzGerald JJ, Antoniades CA. Deep Brain Stimulation and Levodopa Affect Gait Variability in Parkinson Disease Differently. Neuromodulation 2023; 26:382-393. [PMID: 35562261 DOI: 10.1016/j.neurom.2022.04.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/07/2022] [Accepted: 02/06/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Both dopaminergic medication and subthalamic nucleus (STN) deep brain stimulation (DBS) can improve the amplitude and speed of gait in Parkinson disease (PD), but relatively little is known about their comparative effects on gait variability. Gait irregularity has been linked to the degeneration of cholinergic neurons in the pedunculopontine nucleus (PPN). OBJECTIVES The STN and PPN have reciprocal connections, and we hypothesized that STN DBS might improve gait variability by modulating PPN function. Dopaminergic medication should not do this, and we therefore sought to compare the effects of medication and STN DBS on gait variability. MATERIALS AND METHODS We studied 11 patients with STN DBS systems on and off with no alteration to their medication, and 15 patients with PD without DBS systems on and off medication. Participants walked for two minutes in each state, wearing six inertial measurement units. Variability has previously often been expressed in terms of SD or coefficient of variation over a testing session, but these measures conflate long-term variability (eg, gradual slowing, which is not necessarily pathological) with short-term variability (true irregularity). We used Poincaré analysis to separate the short- and long-term variability. RESULTS DBS decreased short-term variability in lower limb gait parameters, whereas medication did not have this effect. In contrast, STN DBS had no effect on arm swing and trunk motion variability, whereas medication increased them, without obvious dyskinesia. CONCLUSIONS Our results suggest that STN DBS acts through a nondopaminergic mechanism to reduce gait variability. We believe that the most likely explanation is the retrograde activation of cholinergic PPN projection neurons.
Collapse
Affiliation(s)
- Zi H Su
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Salil Patel
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Bronwyn Gavine
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Marko Bogdanovic
- Oxford Functional Neurosurgery, John Radcliffe Hospital, Oxford, UK
| | | | - Alexander L Green
- Oxford Functional Neurosurgery, John Radcliffe Hospital, Oxford, UK; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Bastiaan R Bloem
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - James J FitzGerald
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford Functional Neurosurgery, John Radcliffe Hospital, Oxford, UK; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Chrystalina A Antoniades
- NeuroMetrology Lab, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| |
Collapse
|
36
|
Hansen C, Chebil B, Cockroft J, Bianchini E, Romijnders R, Maetzler W. Changes in Coordination and Its Variability with an Increase in Functional Performance of the Lower Extremities. BIOSENSORS 2023; 13:156. [PMID: 36831922 PMCID: PMC9953305 DOI: 10.3390/bios13020156] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
Clinical gait analysis has a long-standing tradition in biomechanics. However, the use of kinematic data or segment coordination has not been reported based on wearable sensors in "real-life" environments. In this work, the skeletal kinematics of 21 healthy and 24 neurogeriatric participants was collected in a magnetically disturbed environment with inertial measurement units (IMUs) using an accelerometer-based functional calibration method. The system consists of seven IMUs attached to the lower back, the thighs, the shanks, and the feet to acquire and process the raw sensor data. The Short Physical Performance Battery (SPPB) test was performed to relate joint kinematics and segment coordination to the overall SPPB score. Participants were then divided into three subgroups based on low (0-6), moderate (7-9), or high (10-12) SPPB scores. The main finding of this study is that most IMU-based parameters significantly correlated with the SPPB score and the parameters significantly differed between the SPPB subgroups. Lower limb range of motion and joint segment coordination correlated positively with the SPPB score, and the segment coordination variability correlated negatively. The results suggest that segment coordination impairments become more pronounced with a decreasing SPPB score, indicating that participants with low overall SPPB scores produce a peculiar inconsistent walking pattern to counteract lower extremity impairment in strength, balance, and mobility. Our findings confirm the usefulness of SPPB through objectively measured parameters, which may be relevant for the design of future studies and clinical routines.
Collapse
Affiliation(s)
- Clint Hansen
- Neurogeriatrics, University Hospital Kiel, 24105 Kiel, Germany
| | - Baraah Chebil
- Neurogeriatrics, University Hospital Kiel, 24105 Kiel, Germany
| | - John Cockroft
- Neuromechanics Unit, Stellenbosch University, Stellenbosch 7602, South Africa
| | | | - Robbin Romijnders
- Neurogeriatrics, University Hospital Kiel, 24105 Kiel, Germany
- Digital Signal Processing and System Theory, Kiel University, 24118 Kiel, Germany
| | - Walter Maetzler
- Neurogeriatrics, University Hospital Kiel, 24105 Kiel, Germany
| |
Collapse
|
37
|
Uhlig M, Prell T. Gait Characteristics Associated with Fear of Falling in Hospitalized People with Parkinson's Disease. SENSORS (BASEL, SWITZERLAND) 2023; 23:1111. [PMID: 36772149 PMCID: PMC9919788 DOI: 10.3390/s23031111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/26/2022] [Accepted: 01/14/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Fear of falling (FOF) is common in Parkinson's disease (PD) and associated with distinct gait changes. Here, we aimed to answer, how quantitative gait assessment can improve our understanding of FOF-related gait in hospitalized geriatric patients with PD. METHODS In this cross-sectional study of 79 patients with advanced PD, FOF was assessed with the Falls Efficacy Scale International (FES-I), and spatiotemporal gait parameters were recorded with a mobile gait analysis system with inertial measurement units at each foot while normal walking. In addition, demographic parameters, disease-specific motor (MDS-revised version of the Unified Parkinson's Disease Rating Scale, Hoehn & Yahr), and non-motor (Non-motor Symptoms Questionnaire, Montreal Cognitive Assessment) scores were assessed. RESULTS According to the FES-I, 22.5% reported low, 28.7% moderate, and 47.5% high concerns about falling. Most concerns were reported when walking on a slippery surface, on an uneven surface, or up or down a slope. In the final regression model, previous falls, more depressive symptoms, use of walking aids, presence of freezing of gait, and lower walking speed explained 42% of the FES-I variance. CONCLUSION Our study suggests that FOF is closely related to gait changes in hospitalized PD patients. Therefore, FOF needs special attention in the rehabilitation of these patients, and targeting distinct gait parameters under varying walking conditions might be a promising part of a multimodal treatment program in PD patients with FOF. The effect of these targeted interventions should be investigated in future trials.
Collapse
Affiliation(s)
- Manuela Uhlig
- Department of Neurology, Jena University Hospital, 07743 Jena, Germany
| | - Tino Prell
- Department of Neurology, Jena University Hospital, 07743 Jena, Germany
- Department of Geriatrics, Halle University Hospital, 06120 Halle, Germany
| |
Collapse
|
38
|
Zhao H, Cao J, Xie J, Liao WH, Lei Y, Cao H, Qu Q, Bowen C. Wearable sensors and features for diagnosis of neurodegenerative diseases: A systematic review. Digit Health 2023; 9:20552076231173569. [PMID: 37214662 PMCID: PMC10192816 DOI: 10.1177/20552076231173569] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/17/2023] [Indexed: 05/24/2023] Open
Abstract
Objective Neurodegenerative diseases affect millions of families around the world, while various wearable sensors and corresponding data analysis can be of great support for clinical diagnosis and health assessment. This systematic review aims to provide a comprehensive overview of the existing research that uses wearable sensors and features for the diagnosis of neurodegenerative diseases. Methods A systematic review was conducted of studies published between 2015 and 2022 in major scientific databases such as Web of Science, Google Scholar, PubMed, and Scopes. The obtained studies were analyzed and organized into the process of diagnosis: wearable sensors, feature extraction, and feature selection. Results The search led to 171 eligible studies included in this overview. Wearable sensors such as force sensors, inertial sensors, electromyography, electroencephalography, acoustic sensors, optical fiber sensors, and global positioning systems were employed to monitor and diagnose neurodegenerative diseases. Various features including physical features, statistical features, nonlinear features, and features from the network can be extracted from these wearable sensors, and the alteration of features toward neurodegenerative diseases was illustrated. Moreover, different kinds of feature selection methods such as filter, wrapper, and embedded methods help to find the distinctive indicator of the diseases and benefit to a better diagnosis performance. Conclusions This systematic review enables a comprehensive understanding of wearable sensors and features for the diagnosis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Huan Zhao
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi'an, P.R. China
| | - Junyi Cao
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi'an, P.R. China
| | - Junxiao Xie
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi'an, P.R. China
| | - Wei-Hsin Liao
- Department of Mechanical and Automation
Engineering, The Chinese University of Hong
Kong, Shatin, N.T., Hong Kong, China
| | - Yaguo Lei
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi'an, P.R. China
| | - Hongmei Cao
- Department of Neurology, The First
Affiliated Hospital of Xi’an Jiaotong University, Xi’an, P.R. China
| | - Qiumin Qu
- Department of Neurology, The First
Affiliated Hospital of Xi’an Jiaotong University, Xi’an, P.R. China
| | - Chris Bowen
- Department of Mechanical Engineering, University of Bath, Bath, UK
| |
Collapse
|
39
|
d'Alencar MS, Santos GV, Helene AF, Roque AC, Miranda JGV, Piemonte MEP. A non-expensive bidimensional assessment can detect subtle alterations in gait performance in people in the early stages of Parkinson's disease. Front Neurol 2023; 14:1101650. [PMID: 37153678 PMCID: PMC10155096 DOI: 10.3389/fneur.2023.1101650] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/15/2023] [Indexed: 05/10/2023] Open
Abstract
Background Gait is one of the activities most affected by the symptoms of Parkinson's disease and may show a linear decline as the disease progresses. Early assessment of its performance through clinically relevant tests is a key factor in designing efficient therapeutic plans and procedures, which can be enhanced using simple and low-cost technological instruments. Objective To investigate the effectiveness of a two-dimensional gait assessment to identify the decline in gait performance associated with Parkinson's disease progression. Methods One hundred and seventeen people with Parkinson's disease, classified between early and intermediate stages, performed three clinical gait tests (Timed Up and Go, Dynamic Gait Index, and item 29 of the Unified Parkinson's Disease Rating Scale), in addition to a six-meter gait test recorded by a two-dimensional movement analysis software. Based on variables generated by the software, a gait performance index was created, allowing a comparison between its results with the results obtained by clinical tests. Results There were differences between sociodemographic variables directly related to the evolution of Parkinson's disease. Compared to clinical tests, the index proposed to analyze gait showed greater sensitivity and was able to differentiate the first three stages of disease evolution (Hoehn and Yahr I and II: p = 0.03; Hoehn and Yahr I and III: p = 0.00001; Hoehn and Yahr II and III: p = 0.02). Conclusion Based on the index provided by a two-dimensional movement analysis software that uses kinematic gait variables, it was possible to differentiate the gait performance decline among the three first stages of Parkinson's disease evolution. This study offers a promising possibility of early identification of subtle changes in an essential function of people with Parkinson's disease.
Collapse
Affiliation(s)
- Matheus Silva d'Alencar
- Department of Physical Therapy, Speech Therapy and Occupational Therapy, Faculty of Medical Science, University of São Paulo, São Paulo, Brazil
| | - Gabriel Venas Santos
- Department of Physical Therapy, Speech Therapy and Occupational Therapy, Faculty of Medical Science, University of São Paulo, São Paulo, Brazil
| | - André Frazão Helene
- Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Antonio Carlos Roque
- Department of Physics, School of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Maria Elisa Pimentel Piemonte
- Department of Physical Therapy, Speech Therapy and Occupational Therapy, Faculty of Medical Science, University of São Paulo, São Paulo, Brazil
- *Correspondence: Maria Elisa Pimentel Piemonte,
| |
Collapse
|
40
|
Geritz J, Welzel J, Hansen C, Maetzler C, Hobert MA, Elshehabi M, Knacke H, Aleknonytė-Resch M, Kudelka J, Bunzeck N, Maetzler W. Cognitive parameters can predict change of walking performance in advanced Parkinson's disease - Chances and limits of early rehabilitation. Front Aging Neurosci 2022; 14:1070093. [PMID: 36620765 PMCID: PMC9813446 DOI: 10.3389/fnagi.2022.1070093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Links between cognition and walking performance in patients with Parkinson's disease (PD), which both decline with disease progression, are well known. There is lack of knowledge regarding the predictive value of cognition for changes in walking performance after individualized therapy. The aim of this study is to identify relevant predictive cognitive and affective parameters, measurable in daily clinical routines, for change in quantitative walking performance after early geriatric rehabilitation. Methods Forty-seven acutely hospitalized patients with advanced PD were assessed at baseline (T1) and at the end (T2) of a 2-week early rehabilitative geriatric complex treatment (ERGCT). Global cognitive performance (Montreal Cognitive Assessment, MoCA), EF and divided attention (Trail Making Test B minus A, delta TMT), depressive symptoms, and fear of falling were assessed at T1. Change in walking performance was determined by the difference in quantitative walking parameters extracted from a sensor-based movement analysis over 20 m straight walking in single (ST, fast and normal pace) and dual task (DT, with secondary cognitive, respectively, motor task) conditions between T1 and T2. Bayesian regression (using Bayes Factor BF10) and multiple linear regression models were used to determine the association of non-motor characteristics for change in walking performance. Results Under ST, there was moderate evidence (BF10 = 7.8, respectively, BF10 = 4.4) that lower performance in the ∆TMT at baseline is associated with lower reduction of step time asymmetry after treatment (R 2 adj = 0.26, p ≤ 0.008, respectively, R 2 adj = 0.18, p ≤ 0.009). Under DT walking-cognitive, there was strong evidence (BF10 = 29.9, respectively, BF10 = 27.9) that lower performance in the ∆TMT is associated with more reduced stride time and double limb support (R 2 adj = 0.62, p ≤ 0.002, respectively, R 2 adj = 0.51, p ≤ 0.009). There was moderate evidence (BF10 = 5.1) that a higher MoCA total score was associated with increased gait speed after treatment (R 2 adj = 0.30, p ≤ 0.02). Discussion Our results indicate that the effect of ERGT on change in walking performance is limited for patients with deficits in EF and divided attention. However, these patients also seem to walk more cautiously after treatment in walking situations with additional cognitive demand. Therefore, future development of individualized treatment algorithms is required, which address individual needs of these vulnerable patients.
Collapse
Affiliation(s)
- Johanna Geritz
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany,Department of Psychology, University of Lübeck, Lübeck, Germany,*Correspondence: Johanna Geritz,
| | - Julius Welzel
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Clint Hansen
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Corina Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Markus A. Hobert
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Morad Elshehabi
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Henrike Knacke
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Jennifer Kudelka
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nico Bunzeck
- Department of Psychology, University of Lübeck, Lübeck, Germany,Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Walter Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
41
|
Scherbaum R, Moewius A, Oppermann J, Geritz J, Hansen C, Gold R, Maetzler W, Tönges L. Parkinson's disease multimodal complex treatment improves gait performance: an exploratory wearable digital device-supported study. J Neurol 2022; 269:6067-6085. [PMID: 35864214 PMCID: PMC9553759 DOI: 10.1007/s00415-022-11257-x] [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: 10/20/2021] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Wearable device-based parameters (DBP) objectively describe gait and balance impairment in Parkinson's disease (PD). We sought to investigate correlations between DBP of gait and balance and clinical scores, their respective changes throughout the inpatient multidisciplinary Parkinson's Disease Multimodal Complex Treatment (PD-MCT), and correlations between their changes. METHODS This exploratory observational study assessed 10 DBP and clinical scores at the start (T1) and end (T2) of a two-week PD-MCT of 25 PD in patients (mean age: 66.9 years, median HY stage: 2.5). Subjects performed four straight walking tasks under single- and dual-task conditions, and four balance tasks. RESULTS At T1, reduced gait velocity and larger sway area correlated with motor severity. Shorter strides during motor-motor dual-tasking correlated with motor complications. From T1 to T2, gait velocity improved, especially under dual-task conditions, stride length increased for motor-motor dual-tasking, and clinical scores measuring motor severity, balance, dexterity, executive functions, and motor complications changed favorably. Other gait parameters did not change significantly. Changes in motor complications, motor severity, and fear of falling correlated with changes in stride length, sway area, and measures of gait stability, respectively. CONCLUSION DBP of gait and balance reflect clinical scores, e.g., those of motor severity. PD-MCT significantly improves gait velocity and stride length and favorably affects additional DBP. Motor complications and fear of falling are factors that may influence the response to PD-MCT. A DBP-based assessment on admission to PD inpatient treatment could allow for more individualized therapy that can improve outcomes. TRIAL REGISTRATION NUMBER AND DATE DRKS00020948 number, 30-Mar-2020, retrospectively registered.
Collapse
Affiliation(s)
- Raphael Scherbaum
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, 44791, Bochum, Germany
| | - Andreas Moewius
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, 44791, Bochum, Germany
| | - Judith Oppermann
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, 44791, Bochum, Germany
| | - Johanna Geritz
- Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Clint Hansen
- Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, 44791, Bochum, Germany
- Neurodegeneration Research, Protein Research Unit Ruhr (PURE), Ruhr University Bochum, 44801, Bochum, Germany
| | - Walter Maetzler
- Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Lars Tönges
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, 44791, Bochum, Germany.
- Neurodegeneration Research, Protein Research Unit Ruhr (PURE), Ruhr University Bochum, 44801, Bochum, Germany.
| |
Collapse
|
42
|
Zhang X, Fan W, Yu H, Li L, Chen Z, Guan Q. Single- and dual-task gait performance and their diagnostic value in early-stage Parkinson's disease. Front Neurol 2022; 13:974985. [PMID: 36313494 PMCID: PMC9615249 DOI: 10.3389/fneur.2022.974985] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Background Gait parameters are considered potential diagnostic markers of Parkinson's disease (PD). We aimed to 1) assess the gait impairment in early-stage PD and its related factors in the single-task (ST) and dual-task (DT) walking tests and 2) evaluate and compare the diagnostic value of gait parameters for early-stage PD under ST and DT conditions. Methods A total of 97 early-stage PD patients and 41 healthy controls (HC) were enrolled at Hwa Mei hospital. Gait parameters were gathered and compared between the two groups in the ST and DT walking test, controlling for covariates. Utilizing the receiver operating characteristic curve, diagnostic parameters were investigated. Results In the ST walking test, significantly altered gait patterns could be observed in early-stage PD patients in all domains of gait, except for asymmetry (P < 0.05). Compared to the ST walking test, the early-stage PD group performed poorly in the DT walking test in the pace, rhythm, variability and postural control domain (P < 0.05). Older, heavier subjects, as well as those with lower height, lower level of education and lower gait velocity, were found to have a poorer gait performance (P < 0.05). Stride length (AUC = 0.823, sensitivity, 68.0%; specificity, 85.4%; P < 0.001) and heel strike angle (AUC = 0.796, sensitivity, 71.1%; specificity, 80.5%; P < 0.001) could distinguish early-stage PD patients from HCs with moderate accuracy, independent of covariates. The diagnostic accuracy of gait parameters under ST conditions were statistically noninferior to those under DT conditions(P>0.05). Combining all gait parameters with diagnostic values under ST and DT walking test, the predictive power significantly increased with an AUC of 0.924 (sensitivity, 85.4%; specificity, 92.7%; P < 0.001). Conclusion Gait patterns altered in patients with early-stage PD but the gait symmetry remained preserved. Stride length and heel strike angle were the two most prominent gait parameters of altered gait in early-stage of PD that could serve as diagnostic markers of early-stage PD. Our findings are helpful to understand the gait pattern of early-stage PD and its related factors and can be conducive to the development of new diagnostic tools for early-stage PD.
Collapse
Affiliation(s)
| | | | | | | | - Zhaoying Chen
- Department of Neurology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Qiongfeng Guan
- Department of Neurology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
43
|
Xu Z, Shen B, Tang Y, Wu J, Wang J. Deep Clinical Phenotyping of Parkinson's Disease: Towards a New Era of Research and Clinical Care. PHENOMICS (CHAM, SWITZERLAND) 2022; 2:349-361. [PMID: 36939759 PMCID: PMC9590510 DOI: 10.1007/s43657-022-00051-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/12/2022] [Accepted: 03/28/2022] [Indexed: 11/27/2022]
Abstract
Despite recent advances in technology, clinical phenotyping of Parkinson's disease (PD) has remained relatively limited as current assessments are mainly based on empirical observation and subjective categorical judgment at the clinic. A lack of comprehensive, objective, and quantifiable clinical phenotyping data has hindered our capacity to diagnose, assess patients' conditions, discover pathogenesis, identify preclinical stages and clinical subtypes, and evaluate new therapies. Therefore, deep clinical phenotyping of PD patients is a necessary step towards understanding PD pathology and improving clinical care. In this review, we present a growing community consensus and perspective on how to clinically phenotype this disease, that is, to phenotype the entire course of disease progression by integrating capacity, performance, and perception approaches with state-of-the-art technology. We also explore the most studied aspects of PD deep clinical phenotypes, namely, bradykinesia, tremor, dyskinesia and motor fluctuation, gait impairment, speech impairment, and non-motor phenotypes.
Collapse
Affiliation(s)
- Zhiheng Xu
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Bo Shen
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Yilin Tang
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Jianjun Wu
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Jian Wang
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| |
Collapse
|
44
|
Tam W, Alajlani M, Abd-alrazaq A. An Exploration of Wearable Device Features Used in UK Hospital Parkinson Disease Care: Scoping Review (Preprint).. [DOI: 10.2196/preprints.42950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
BACKGROUND
The prevalence of Parkinson disease (PD) is becoming an increasing concern owing to the aging population in the United Kingdom. Wearable devices have the potential to improve the clinical care of patients with PD while reducing health care costs. Consequently, exploring the features of these wearable devices is important to identify the limitations and further areas of investigation of how wearable devices are currently used in clinical care in the United Kingdom.
OBJECTIVE
In this scoping review, we aimed to explore the features of wearable devices used for PD in hospitals in the United Kingdom.
METHODS
A scoping review of the current research was undertaken and reported according to the PRISMA-ScR (Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews) guidelines. The literature search was undertaken on June 6, 2022, and publications were obtained from MEDLINE or PubMed, Embase, and the Cochrane Library. Eligible publications were initially screened by their titles and abstracts. Publications that passed the initial screening underwent a full review. The study characteristics were extracted from the final publications, and the evidence was synthesized using a narrative approach. Any queries were reviewed by the first and second authors.
RESULTS
Of the 4543 publications identified, 39 (0.86%) publications underwent a full review, and 20 (0.44%) publications were included in the scoping review. Most studies (11/20, 55%) were conducted at the Newcastle upon Tyne Hospitals NHS Foundation Trust, with sample sizes ranging from 10 to 418. Most study participants were male individuals with a mean age ranging from 57.7 to 78.0 years. The AX3 was the most popular device brand used, and it was commercially manufactured by Axivity. Common wearable device types included body-worn sensors, inertial measurement units, and smartwatches that used accelerometers and gyroscopes to measure the clinical features of PD. Most wearable device primary measures involved the measured gait, bradykinesia, and dyskinesia. The most common wearable device placements were the lumbar region, head, and wrist. Furthermore, 65% (13/20) of the studies used artificial intelligence or machine learning to support PD data analysis.
CONCLUSIONS
This study demonstrated that wearable devices could help provide a more detailed analysis of PD symptoms during the assessment phase and personalize treatment. Using machine learning, wearable devices could differentiate PD from other neurodegenerative diseases. The identified evidence gaps include the lack of analysis of wearable device cybersecurity and data management. The lack of cost-effectiveness analysis and large-scale participation in studies resulted in uncertainty regarding the feasibility of the widespread use of wearable devices. The uncertainty around the identified research gaps was further exacerbated by the lack of medical regulation of wearable devices for PD, particularly in the United Kingdom where regulations were changing due to the political landscape.
CLINICALTRIAL
Collapse
|
45
|
Zhu S, Wu Z, Wang Y, Jiang Y, Gu R, Zhong M, Jiang X, Shen B, Zhu J, Yan J, Pan Y, Zhang L. Gait Analysis with Wearables Is a Potential Progression Marker in Parkinson's Disease. Brain Sci 2022; 12:1213. [PMID: 36138949 PMCID: PMC9497215 DOI: 10.3390/brainsci12091213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/17/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Gait disturbance is a prototypical feature of Parkinson's disease (PD), and the quantification of gait using wearable sensors is promising. This study aimed to identify gait impairment in the early and progressive stages of PD according to the Hoehn and Yahr (H-Y) scale. A total of 138 PD patients and 56 healthy controls (HCs) were included in our research. We collected gait parameters using the JiBuEn gait-analysis system. For spatiotemporal gait parameters and kinematic gait parameters, we observed significant differences in stride length (SL), gait velocity, the variability of SL, heel strike angle, and the range of motion (ROM) of the ankle, knee, and hip joints between HCs and PD patients in H-Y Ⅰ-Ⅱ. The changes worsened with the progression of PD. The differences in the asymmetry index of the SL and ROM of the hip were found between HCs and patients in H-Y Ⅳ. Additionally, these gait parameters were significantly associated with Unified Parkinson's Disease Rating Scale and Parkinson's Disease Questionnaire-39. This study demonstrated that gait impairment occurs in the early stage of PD and deteriorates with the progression of the disease. The gait parameters mentioned above may help to detect PD earlier and assess the progression of PD.
Collapse
Affiliation(s)
- Sha Zhu
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhuang Wu
- Neurotoxin Research Center of Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Neurological Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yaxi Wang
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yinyin Jiang
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ruxin Gu
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Min Zhong
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xu Jiang
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Bo Shen
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jun Zhu
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jun Yan
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yang Pan
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Li Zhang
- Department of Geriatric Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
46
|
Lin F, Yang B, Chen Y, Zhao W, Li B, Jia W. Enlarged perivascular spaces are linked to freezing of gait in Parkinson's disease. Front Neurol 2022; 13:985294. [PMID: 36062021 PMCID: PMC9437541 DOI: 10.3389/fneur.2022.985294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/03/2022] [Indexed: 11/29/2022] Open
Abstract
Objective Freezing of gait (FOG) is one of common and disabling gait impairments of Parkinson's disease (PD). White matter hyperintensity (WMH) and lacunes, as common manifestations of cerebral small vessel diseases (CSVD), have been reported to be associated with gait function in PD patients. However, in the cases with FOG which present with extensive WMH or lacunes, it actually is difficult to distinguish pure PD pathology from vascular origin or combined effects. So far little is known about the correlation between enlarged perivascular space (PVS) and FOG in PD patients. This study aims to explore the role of enlarged PVS in FOG in PD patients. Methods A total of 95 patients with PD in the absence of obvious WMH and lacunes were included in our study, which were divided into PD-FOG (+) group and PD-FOG (-) group. Demographic and clinical data were investigated. Enlarged PVS in the centrum semiovale (CSO) and basal ganglia (BG) were assessed. The association between enlarged PVS and FOG in patients with PD was analyzed using the multivariate models and the Spearman's correlation. Results There were 36 PD patients grouped into PD-FOG (+) (37.9%), with an older age, a longer PD disease duration, and larger numbers of enlarged PVS in CSO and BG compared with PD-FOG (-) group. The highest-severity degree of enlarged PVS burden in CSO was independently associated with FOG in patients with PD [adjusted odds ratio (OR), 3.869; p = 0.022 in multivariable model]. The percentages of FOG case increased accompanied by the aggravation of enlarged PVS located in CSO. The grade and count of enlarged PVS in CSO and BG both correlated with FOGQ score in PD patients. Conclusion Enlarged PVS, particularly in CSO, are associated with FOG in patients with PD, which provides a novel perspective for the mechanisms of FOG in PD.
Collapse
|
47
|
Molsberry SA, Hughes KC, Schwarzschild MA, Ascherio A. Who to Enroll in Parkinson Disease Prevention Trials? The Case for Composite Prodromal Cohorts. Neurology 2022; 99:26-33. [PMID: 35970591 DOI: 10.1212/wnl.0000000000200788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/11/2022] [Indexed: 11/15/2022] Open
Abstract
Significant progress has been made in expanding our understanding of prodromal Parkinson disease (PD), particularly for recognition of early motor and nonmotor signs and symptoms. Although identification of these prodromal features may improve our understanding of the earliest stages of PD, they are individually insufficient for early disease detection and enrollment of participants in prevention trials in most cases because of low sensitivity, specificity, and positive predictive value. Composite cohorts, composed of individuals with multiple co-occurring prodromal features, are an important resource for conducting prodromal PD research and eventual prevention trials because they are more representative of the population at risk for PD, allow investigators to evaluate the efficacy of an intervention across individuals with varying prodromal feature patterns, are able to produce larger sample sizes, and capture individuals at different stages of prodromal PD. A key challenge in identifying individuals with prodromal disease for composite cohorts and prevention trial participation is that we know little about the natural history of prodromal PD. To move toward prevention trials, it is critical that we better understand common prodromal feature patterns and be able to predict the probability of progression and phenoconversion. Ongoing research in cohort studies and administrative databases is beginning to address these questions, but further longitudinal analyses in a large population-based sample are necessary to provide a convincing and definitive strategy for identifying individuals to be enrolled in a prevention trial.
Collapse
Affiliation(s)
- Samantha A Molsberry
- From the Department of Nutrition (S.A.M., A.A.), Harvard T.H. Chan School of Public Health; Epidemiology (K.C.H.), Optum; Department of Neurology (M.A.S.), and MassGeneral Institute for Neurodegenerative Disease (M.A.S.), Massachusetts General Hospital; Department of Epidemiology (A.A.), Harvard T.H. Chan School of Public Health; and Channing Division of Network Medicine (A.A.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA.
| | - Katherine C Hughes
- From the Department of Nutrition (S.A.M., A.A.), Harvard T.H. Chan School of Public Health; Epidemiology (K.C.H.), Optum; Department of Neurology (M.A.S.), and MassGeneral Institute for Neurodegenerative Disease (M.A.S.), Massachusetts General Hospital; Department of Epidemiology (A.A.), Harvard T.H. Chan School of Public Health; and Channing Division of Network Medicine (A.A.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Michael A Schwarzschild
- From the Department of Nutrition (S.A.M., A.A.), Harvard T.H. Chan School of Public Health; Epidemiology (K.C.H.), Optum; Department of Neurology (M.A.S.), and MassGeneral Institute for Neurodegenerative Disease (M.A.S.), Massachusetts General Hospital; Department of Epidemiology (A.A.), Harvard T.H. Chan School of Public Health; and Channing Division of Network Medicine (A.A.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Alberto Ascherio
- From the Department of Nutrition (S.A.M., A.A.), Harvard T.H. Chan School of Public Health; Epidemiology (K.C.H.), Optum; Department of Neurology (M.A.S.), and MassGeneral Institute for Neurodegenerative Disease (M.A.S.), Massachusetts General Hospital; Department of Epidemiology (A.A.), Harvard T.H. Chan School of Public Health; and Channing Division of Network Medicine (A.A.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
48
|
Mirelman A, Siderowf A, Chahine L. Outcome Assessment in Parkinson Disease Prevention Trials: Utility of Clinical and Digital Measures. Neurology 2022; 99:52-60. [PMID: 35970590 DOI: 10.1212/wnl.0000000000200236] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/21/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The prodromal phase of Parkinson disease (PD) is accompanied by subtle clinical signs that are not sufficient for diagnosis but could potentially be measured in the context of clinical trials of therapies intended to delay or prevent more definitive clinical features. The objective of this study was to review the available literature on the presence and time course of subtle motor features in prodromal PD in the context of planning for possible clinical trials. METHODS We reviewed the available literature based on expert opinion. We considered a range of outcomes including measurement of clinical features, patient-reported outcomes, digital markers, and clinical diagnosis. RESULTS We considered these features and measures in the context of patient stratification, intermediate outcomes, and clinically relevant end points, including phenoconversion. DISCUSSION Substantial progress has been made in understanding how motor features evolve in the period immediately before a PD diagnosis. Digital measures hold substantial progress for measurement precision and may be additionally relevant because they can be used in naturalistic environments outside the clinic. Future studies should focus on advancing digital sensor technology and analysis and developing methods to implement available methods, particularly determination of a clinical diagnosis of PD, in a clinical trial context.
Collapse
Affiliation(s)
- Anat Mirelman
- From the Sackler School of Medicine and Sagol School of Neuroscience (A.M.), Tel Aviv University, Israel; Department of Neurology (A.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Neurology (L.C.), University of Pittsburgh, PA
| | - Andrew Siderowf
- From the Sackler School of Medicine and Sagol School of Neuroscience (A.M.), Tel Aviv University, Israel; Department of Neurology (A.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Neurology (L.C.), University of Pittsburgh, PA.
| | - Lana Chahine
- From the Sackler School of Medicine and Sagol School of Neuroscience (A.M.), Tel Aviv University, Israel; Department of Neurology (A.S.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; and Department of Neurology (L.C.), University of Pittsburgh, PA
| |
Collapse
|
49
|
Berg D, Crotty GF, Keavney JL, Schwarzschild MA, Simuni T, Tanner C. Path to Parkinson Disease Prevention: Conclusion and Outlook. Neurology 2022; 99:76-83. [PMID: 35970586 DOI: 10.1212/wnl.0000000000200793] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/12/2022] [Indexed: 01/19/2023] Open
Abstract
Tremendous progress in our understanding of the pathophysiology and clinical manifestations of the prodromal phase of Parkinson disease (PD) offers a unique opportunity to start therapeutic interventions as early as possible to slow or even stop the progression to clinically manifest motor PD. A Parkinson's Prevention Conference, "Planning for Prevention of Parkinson's: A trial design symposium and workshop" was convened to discuss all issues that need to be addressed before the launch of the first PD prevention study. In this review, we summarize the major opportunities and challenges in designing prevention trials in PD, organized by the following critical trial design questions: Who (should be enrolled)? What (to test)? How (to measure prevention)? and the pivotal question, When during the prodromal disease (should we start these trials)? We outline the implications of these questions and their meaning for a responsible, sustainable, and fruitful further planning for prevention trials. Despite the great progress that has been made, it needs to be acknowledged that several queries remain to be carefully considered and addressed because prevention trials are being planned and become a reality.
Collapse
Affiliation(s)
- Daniela Berg
- From the Department of Neurology (D.B.), Christian-Albrechts-University, Kiel, Germany; Molecular Neurobiology Laboratory (G.F.C., M.A.S.), Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown; Harvard Medical School (G.F.C., M.A.S.), Boston, MA; Parkinson's Foundation Research Advocates Program (J.L.K.), Parkinson's Foundation, Miami, FL/New York, NY; Northwestern University Feinberg School of Medicine (T.S.), Weill Institute for Neuroscience (C.T.), Department of Neurology, University of California - San Francisco; and Parkinson's Disease Research Education and Clinical Center (C.T.), San Francisco Veterans Affairs Medical Center
| | - Grace F Crotty
- From the Department of Neurology (D.B.), Christian-Albrechts-University, Kiel, Germany; Molecular Neurobiology Laboratory (G.F.C., M.A.S.), Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown; Harvard Medical School (G.F.C., M.A.S.), Boston, MA; Parkinson's Foundation Research Advocates Program (J.L.K.), Parkinson's Foundation, Miami, FL/New York, NY; Northwestern University Feinberg School of Medicine (T.S.), Weill Institute for Neuroscience (C.T.), Department of Neurology, University of California - San Francisco; and Parkinson's Disease Research Education and Clinical Center (C.T.), San Francisco Veterans Affairs Medical Center
| | - Jessi L Keavney
- From the Department of Neurology (D.B.), Christian-Albrechts-University, Kiel, Germany; Molecular Neurobiology Laboratory (G.F.C., M.A.S.), Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown; Harvard Medical School (G.F.C., M.A.S.), Boston, MA; Parkinson's Foundation Research Advocates Program (J.L.K.), Parkinson's Foundation, Miami, FL/New York, NY; Northwestern University Feinberg School of Medicine (T.S.), Weill Institute for Neuroscience (C.T.), Department of Neurology, University of California - San Francisco; and Parkinson's Disease Research Education and Clinical Center (C.T.), San Francisco Veterans Affairs Medical Center
| | - Michael A Schwarzschild
- From the Department of Neurology (D.B.), Christian-Albrechts-University, Kiel, Germany; Molecular Neurobiology Laboratory (G.F.C., M.A.S.), Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown; Harvard Medical School (G.F.C., M.A.S.), Boston, MA; Parkinson's Foundation Research Advocates Program (J.L.K.), Parkinson's Foundation, Miami, FL/New York, NY; Northwestern University Feinberg School of Medicine (T.S.), Weill Institute for Neuroscience (C.T.), Department of Neurology, University of California - San Francisco; and Parkinson's Disease Research Education and Clinical Center (C.T.), San Francisco Veterans Affairs Medical Center
| | - Tanya Simuni
- From the Department of Neurology (D.B.), Christian-Albrechts-University, Kiel, Germany; Molecular Neurobiology Laboratory (G.F.C., M.A.S.), Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown; Harvard Medical School (G.F.C., M.A.S.), Boston, MA; Parkinson's Foundation Research Advocates Program (J.L.K.), Parkinson's Foundation, Miami, FL/New York, NY; Northwestern University Feinberg School of Medicine (T.S.), Weill Institute for Neuroscience (C.T.), Department of Neurology, University of California - San Francisco; and Parkinson's Disease Research Education and Clinical Center (C.T.), San Francisco Veterans Affairs Medical Center.
| | - Caroline Tanner
- From the Department of Neurology (D.B.), Christian-Albrechts-University, Kiel, Germany; Molecular Neurobiology Laboratory (G.F.C., M.A.S.), Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown; Harvard Medical School (G.F.C., M.A.S.), Boston, MA; Parkinson's Foundation Research Advocates Program (J.L.K.), Parkinson's Foundation, Miami, FL/New York, NY; Northwestern University Feinberg School of Medicine (T.S.), Weill Institute for Neuroscience (C.T.), Department of Neurology, University of California - San Francisco; and Parkinson's Disease Research Education and Clinical Center (C.T.), San Francisco Veterans Affairs Medical Center
| |
Collapse
|
50
|
Geritz J, Welzel J, Hansen C, Maetzler C, Hobert MA, Elshehabi M, Sobczak A, Kudelka J, Stiel C, Hieke J, Alpes A, Bunzeck N, Maetzler W. Does Executive Function Influence Walking in Acutely Hospitalized Patients With Advanced Parkinson's Disease: A Quantitative Analysis. Front Neurol 2022; 13:852725. [PMID: 35928127 PMCID: PMC9344922 DOI: 10.3389/fneur.2022.852725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionIt is well-known that, in Parkinson's disease (PD), executive function (EF) and motor deficits lead to reduced walking performance. As previous studies investigated mainly patients during the compensated phases of the disease, the aim of this study was to investigate the above associations in acutely hospitalized patients with PD.MethodsA total of seventy-four acutely hospitalized patients with PD were assessed with the delta Trail Making Test (ΔTMT, TMT-B minus TMT-A) and the Movement Disorder Society-revised version of the motor part of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS III). Walking performance was assessed with wearable sensors under single (ST; fast and normal pace) and dual-task (DT; walking and checking boxes as the motor secondary task and walking and subtracting seven consecutively from a given three-digit number as the cognitive secondary task) conditions over 20 m. Multiple linear regression and Bayes factor BF10 were performed for each walking parameter and their dual-task costs while walking (DTC) as dependent variables and also included ΔTMT, MDS-UPDRS III, age, and gender.ResultsUnder ST, significant negative effects of the use of a walking aid and MDS-UPDRS III on gait speed and at a fast pace on the number of steps were observed. Moreover, depending on the pace, the use of a walking aid, age, and gender affected step time variability. Under walking-cognitive DT, a resolved variance of 23% was observed in the overall model for step time variability DTC, driven mainly by age (β = 0.26, p = 0.09). Under DT, no other significant effects could be observed. ΔTMT showed no significant associations with any of the walking conditions.DiscussionThe results of this study suggest that, in acutely hospitalized patients with PD, reduced walking performance is mainly explained by the use of a walking aid, motor symptoms, age, and gender, and EF deficits surprisingly do not seem to play a significant role. However, these patients with PD should avoid walking-cognitive DT situations, as under this condition, especially step time variability, a parameter associated with the risk of falling in PD worsens.
Collapse
Affiliation(s)
- Johanna Geritz
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Psychology and Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- *Correspondence: Johanna Geritz
| | - Julius Welzel
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Clint Hansen
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Corina Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Markus A. Hobert
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Morad Elshehabi
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Alexandra Sobczak
- Department of Psychology and Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Jennifer Kudelka
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christopher Stiel
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Johanne Hieke
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Annekathrin Alpes
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nico Bunzeck
- Department of Psychology and Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Walter Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|