1
|
Lee I, Mitsumoto H, Lee S, Kasarskis E, Rosenbaum M, Factor-Litvak P, Nieves JW. Higher Glycemic Index and Glycemic Load Diet Is Associated with Slower Disease Progression in Amyotrophic Lateral Sclerosis. Ann Neurol 2024; 95:217-229. [PMID: 37975189 PMCID: PMC10842093 DOI: 10.1002/ana.26825] [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: 06/18/2023] [Revised: 09/23/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVE High-caloric diets may slow the progression of amyotrophic lateral sclerosis; however, key macronutrients have not been identified. We examined whether dietary macronutrients are associated with the rate of progression and length of survival among the prospective cohort study participants. METHODS Participants with a confirmed diagnosis of sporadic amyotrophic lateral sclerosis enrolled in the Multicenter Cohort Study of Oxidative Stress were included (n = 304). We evaluated baseline macronutrient intake assessed by food frequency questionnaire in relation to change in revised amyotrophic lateral sclerosis functional rating scale total-score, and tracheostomy-free survival using linear regression and Cox proportional hazard models. Baseline age, sex, disease duration, diagnostic certainty, body mass index, bulbar onset, revised amyotrophic lateral sclerosis functional rating scale total-score, and forced vital capacity were included as covariates. RESULTS Baseline higher glycemic index and load were associated with less decline of revised amyotrophic lateral sclerosis functional rating scale total score at 3-month follow-up (β = -0.13, 95% CI -0.2, -0.01, p = 0.03) and (β = -0.01, 95% CI -0.03, -0.0007, p = 0.04), respectively. Glycemic index second-quartile, third-quartile, and fourth-quartile groups were associated with less decline at 3 months by 1.9 (95% CI -3.3, -0.5, p = 0.008), 2.0 (95% CI -3.3, -0.6, p = 0.006), and 1.6 (95% CI -3.0, -0.2, p = 0.03) points compared with the first-quartile group; the glycemic load fourth-quartile group had 1.4 points less decline compared with the first-quartile group (95% CI -2.8, 0.1, p = 0.07). Higher glycemic index was associated with a trend toward longer tracheostomy-free survival (HR 0.97, 95% CI 0.93, 1.00, p = 0.07). INTERPRETATION Higher dietary glycemic index and load are associated with slower disease progression in amyotrophic lateral sclerosis. ANN NEUROL 2024;95:217-229.
Collapse
Affiliation(s)
- Ikjae Lee
- Department of Neurology Columbia University Irving Medical Center, New York, NY
| | - Hiroshi Mitsumoto
- Department of Neurology Columbia University Irving Medical Center, New York, NY
| | - Seonjoo Lee
- Department of Biostatistics and Psychiatry, Columbia University, New York, NY
- Mental Health Data Science, New York State Psychiatric Institute, New York, NY
| | | | - Michael Rosenbaum
- Department of Pediatrics and Medicine, Columbia University Irving Medical Center, New York, NY
| | - Pam Factor-Litvak
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY
| | - Jeri W. Nieves
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY
| |
Collapse
|
2
|
Pottinger TD, Motelow JE, Povysil G, Moreno CAM, Ren Z, Phatnani H, Aitman TJ, Santoyo-Lopez J, Mitsumoto H, Goldstein DB, Harms MB. Rare variant analyses validate known ALS genes in a multi-ethnic population and identifies ANTXR2 as a candidate in PLS. RESEARCH SQUARE 2023:rs.3.rs-3721598. [PMID: 38196621 PMCID: PMC10775375 DOI: 10.21203/rs.3.rs-3721598/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting over 30,000 people in the United States. It is characterized by the progressive decline of the nervous system that leads to the weakening of muscles which impacts physical function. Approximately, 15% of individuals diagnosed with ALS have a known genetic variant that contributes to their disease. As therapies that slow or prevent symptoms, such as antisense oligonucleotides, continue to develop, it is important to discover novel genes that could be targets for treatment. Additionally, as cohorts continue to grow, performing analyses in ALS subtypes, such as primary lateral sclerosis (PLS), becomes possible due to an increase in power. These analyses could highlight novel pathways in disease manifestation. Methods Building on our previous discoveries using rare variant association analyses, we conducted rare variant burden testing on a substantially larger cohort of 6,970 ALS patients from a large multi-ethnic cohort as well as 166 PLS patients, and 22,524 controls. We used intolerant domain percentiles based on sub-region Residual Variation Intolerance Score (subRVIS) that have been described previously in conjunction with gene based collapsing approaches to conduct burden testing to identify genes that associate with ALS and PLS. Results A gene based collapsing model showed significant associations with SOD1, TARDBP, and TBK1 (OR=19.18, p = 3.67 × 10-39; OR=4.73, p = 2 × 10-10; OR=2.3, p = 7.49 × 10-9, respectively). These genes have been previously associated with ALS. Additionally, a significant novel control enriched gene, ALKBH3 (p = 4.88 × 10-7), was protective for ALS in this model. An intolerant domain based collapsing model showed a significant improvement in identifying regions in TARDBP that associated with ALS (OR=10.08, p = 3.62 × 10-16). Our PLS protein truncating variant collapsing analysis demonstrated significant case enrichment in ANTXR2 (p=8.38 × 10-6). Conclusions In a large multi-ethnic cohort of 6,970 ALS patients, rare variant burden testing validated known ALS genes and identified a novel potentially protective gene, ALKBH3. A first-ever analysis in 166 patients with PLS found a candidate association with loss-of-function mutations in ANTXR2.
Collapse
Affiliation(s)
- Tess D. Pottinger
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Internal Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Joshua E. Motelow
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Gundula Povysil
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | | | - Zhong Ren
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Hemali Phatnani
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, United States of America
- Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, New York, United States of America
- New York Genome Center, New York, New York, United States of America
| | | | - Timothy J. Aitman
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
| | | | | | - Hiroshi Mitsumoto
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, United States of America
| | | | - David B. Goldstein
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Matthew B. Harms
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, United States of America
- Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, New York, United States of America
| |
Collapse
|
3
|
Boyle J, Wheeler DC, Naum R, Burke Brockenbrough P, Gebhardt M, Smith L, Harrell T, Stewart D, Gwathmey K. Analysis of the spatial distribution of amyotrophic lateral sclerosis in Virginia. Amyotroph Lateral Scler Frontotemporal Degener 2023:1-9. [PMID: 37452450 DOI: 10.1080/21678421.2023.2236653] [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: 03/20/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
Objective: Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that is usually fatal. Environmental exposures have been posited in the etiology of ALS, but few studies have modeled the spatial risk of ALS over large geographic areas. In this paper, our goal was to analyze the spatial distribution of ALS in Virginia and identify any areas with significantly elevated risk using Virginia ALS Association administrative data. Methods: We used Bayesian hierarchical spatial regression models to estimate the relative risk for ALS in Virginia census tracts, adjusting for several covariates posited to be associated with the disease. We used an intrinsic conditional autoregressive prior to allow for spatial correlation in the risk estimates and stabilize estimates over space. Results: Considerable variation in ALS risk existed across Virginia, with greater relative risk found in the central and western parts of the state. We identified significantly elevated relative risk in a number of census tracts. In particular, Henrico, Albemarle, and Botetourt counties all contained at least four census tracts with significantly elevated risk. Conclusions: We identified several areas with significantly elevated ALS risk across Virginia census tracts. These results can inform future studies of potential environmental triggers for the disease, whose etiology is still being understood.
Collapse
Affiliation(s)
- Joseph Boyle
- Department of Biostatistics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - David C Wheeler
- Department of Biostatistics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Ryan Naum
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA, and
| | - Paula Burke Brockenbrough
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA, and
| | - Michelle Gebhardt
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA, and
| | - LaVon Smith
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA, and
| | | | | | - Kelly Gwathmey
- Department of Neurology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA, and
| |
Collapse
|
4
|
Bireley JD, Morren JA. CNM-Au8: an experimental agent for the treatment of amyotrophic lateral sclerosis (ALS). Expert Opin Investig Drugs 2023; 32:677-683. [PMID: 37642362 DOI: 10.1080/13543784.2023.2252738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/21/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION Two established disease-specific therapies for the treatment of amyotrophic lateral sclerosis (ALS) are riluzole and edaravone. Limitations of these medications include minimal progression slowing or survival benefit, and effectiveness only in selected populations, particularly for edaravone. AMX0035 and tofersen received US FDA approval in September 2022 and April 2023, respectively. However, phase 3 trials, further examining both medications' efficacy, are ongoing. CNM-Au8 is an efficient catalyst of energy metabolism and is therefore a potential disease-modifying treatment for ALS, a neurodegenerative condition in which there is bioenergetics impairment. AREAS COVERED In this review, we provide an overview of the current ALS treatment market, followed by a description of the pharmacodynamics and pharmacokinetics of CNM-Au8. The main preclinical and available early clinical evidence of CNM-Au8 is then described, as well as its potential as an ALS treatment. EXPERT OPINION Oral treatment with CNM-Au8 failed to meet primary clinical and electrodiagnostic endpoints in phase 2/3 clinical trials. Despite this failure, a number of exploratory endpoints included in phase 2/3 trials suggest CNM-Au8 has the potential to significantly slow clinical worsening, improve quality of life, and prolong survival in ALS. Further study of CNM-Au8 in a phase 3 clinical trial is currently underway.
Collapse
Affiliation(s)
- J Daniel Bireley
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - John A Morren
- Neuromuscular Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
5
|
Lee I, Stingone JA, Chan RB, Mitsumoto H. Utilizing machine learning and lipidomics to distinguish primary lateral sclerosis from amyotrophic lateral sclerosis. Muscle Nerve 2023; 67:306-310. [PMID: 36747323 DOI: 10.1002/mus.27797] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023]
Abstract
INTRODUCTION/AIMS There are currently no imaging or blood diagnostic biomarkers that can differentiate amyotrophic lateral sclerosis (ALS) from primary lateral sclerosis (PLS) patients early in their disease courses. Our objective is to examine whether patients with PLS can be differentiated from ALS reliably by using plasma lipidome profile and supervised machine learning. METHODS 40 ALS and 28 PLS patients derived from the Multicenter Cohort study of Oxidative Stress (COSMOS) and 28 healthy control volunteers (CTR) were included. ALS, PLS, and CTR were matched by age and sex. Plasma samples were obtained after overnight fasting. Lipids were extracted from the plasma samples and analyzed using liquid chromatography/mass spectrometry to obtain relative concentrations of 392 lipid species. The lipid data were partitioned into training and testing datasets randomly. An elastic net algorithm was trained using cross-validation to classify PLS vs ALS and PLS vs CTR. Final accuracy was evaluated in the testing dataset. RESULTS The elastic net model trained with labeled PLS and ALS training lipid dataset demonstrated accuracy (number classified correctly/total number), sensitivity, and specificity of 100% in classifying PLS vs ALS in the unlabeled testing lipid dataset. Similarly, the elastic net model trained with labeled PLS and CTR training lipid datasets demonstrated accuracy, sensitivity, and specificity of 88% in classifying PLS vs CTR in the unlabeled testing lipid dataset. DISCUSSION Our study suggests PLS patients can be accurately distinguished from ALS and CTR by combining lipidome profile and supervised machine learning without clinical information.
Collapse
Affiliation(s)
- Ikjae Lee
- Department of Neurology, Columbia University, New York, New York, USA
| | - Jeanette A Stingone
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Robin Barry Chan
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Hiroshi Mitsumoto
- Department of Neurology, Columbia University, New York, New York, USA
| |
Collapse
|
6
|
Mitsumoto H, Jang G, Lee I, Simmons Z, Sherman AV, Heitzman D, Sorenson E, Cheung K, Andrews J, Harms M, Shneider NA, Santella R, Paganoni S, Ajroud-Driss S, Fernandes JAM, Burke KM, Gwathmey K, Habib AA, Maragakis NJ, Walk D, Fournier C, Heiman-Patterson T, Wymer J, Diaz F, Scelsa SN, Elman L, Genge A, Goutman SA, Hayat G, Jawdat O, Johnston WS, Joyce NC, Kasarskis EJ, Kisanuki YY, Lomen-Hoerth C, Pulley MT, Shah JS, Shoesmith C, Zinman L. Primary lateral sclerosis natural history study - planning, designing, and early enrollment. Amyotroph Lateral Scler Frontotemporal Degener 2022:1-11. [PMID: 36576200 DOI: 10.1080/21678421.2022.2161912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction/Aims. Primary lateral sclerosis (PLS) is exceedingly rare and has been an enigmatic disease. Recent progress has drastically changed this perception, with early biomarkers being investigated and potential medications for PLS emerging at the preclinical stage. The aim of this paper is to describe a study of PLS natural history and discuss the limitations and proposed solutions to the study of a rare and slowly progressive disease. Methods. The PLS Natural History Study is a 30-site, 24-month, prospective study that is supported by multiple funding sources. The study aims to enroll 50 early PLS (disease duration ≤4 years) and 50 definite PLS (disease duration 4 to 15 years) participants using modified PLS Diagnostic Criteria. Smartphone-based assessments including semi-quantitative and quantitative measures and patient-reported outcomes are utilized. In-person quantitative measures are also completed during site visits. The change in the PLS Functional Rating Scale score is the primary outcome. The study utilizes the NeuroBANK® patient-centric data capture and management platform. The biostatistical analysis plan has been developed. Results. In one year, 28 participants have been recruited. Enrollment has been much slower than anticipated due to the COVID-19 pandemic, the rarity of PLS, and potential study competition for internal resources from ALS clinical trials. Discussion. We discuss the need for more innovative methods to enroll and study individuals with such rare diseases and propose a number of mechanisms by which more efficient enrollment could be facilitated.
Collapse
Affiliation(s)
- Hiroshi Mitsumoto
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Grace Jang
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ikjae Lee
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Zachary Simmons
- Department of Neurology, Penn State University College of Medicine, Hershey, PA, USA
| | - Alexander V Sherman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Eric Sorenson
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Ken Cheung
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jinsy Andrews
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Matthew Harms
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Neil A Shneider
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Regina Santella
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Sabrina Paganoni
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | | | - J Americo M Fernandes
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Katherine M Burke
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Kelly Gwathmey
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, USA
| | - Ali A Habib
- Department of Neurology, University of California, Irvine, Orange, CA, USA
| | - Nicholas J Maragakis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David Walk
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | | | | | - James Wymer
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Frank Diaz
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen N Scelsa
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren Elman
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angela Genge
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | - Ghazala Hayat
- Department of Neurology, Saint Louis University, Saint Louis, MO, USA
| | - Omar Jawdat
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Wendy S Johnston
- Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Nanette C Joyce
- Department of Physical Medicine and Rehabilitation, University of California, Davis, Sacramento, CA, USA
| | | | - Yaz Y Kisanuki
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Michael T Pulley
- Department of Neurology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Jaimin S Shah
- Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | | | - Lorne Zinman
- Department of Neurology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | |
Collapse
|
7
|
Modeling of mutant superoxide dismutase 1 octamers with cross-linked disulfide bonds. J Mol Model 2022; 28:89. [DOI: 10.1007/s00894-022-05072-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 02/26/2022] [Indexed: 10/18/2022]
|
8
|
Chavda V, Patel C, Modh D, Ertas YN, Sonak SS, Munshi NK, Anand K, Soni A, Pandey S. Therapeutic Approaches to Amyotrophic Lateral Sclerosis from the Lab to the Clinic. Curr Drug Metab 2022; 23:200-222. [PMID: 35272595 DOI: 10.2174/1389200223666220310113110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 01/07/2022] [Accepted: 02/02/2022] [Indexed: 11/22/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a terminal neuro-degenerative disorder that is clinically recognized as a gradual degeneration of the upper and lower motor neurons, with an average duration of 3 to 5 years from initiation of symptoms to death. The mechanisms underlying the pathogenesis and progression of the disease are multifactorial. Therefore, to find effective treatments, it is necessary to understand this heterogeneity underlying the progression of ALS. Recent developments in gene therapy have opened a new avenue to treat this condition, especially for the characterized genetic types. Gene therapy methods have been studied in a variety of pre-clinical settings and clinical trials, and they may be a promising path for developing an effective and safe ALS cure. A growing body of evidence demonstrates abnormalities in energy metabolism at the cellular and whole-body level in animal models and in people living with ALS. The use and incorporation of high-throughput "omics" methods has radically transformed our thought about ALS, strengthening our understanding of the disease's dynamic molecular architecture, differentiating distinct patient subtypes, and creating a reasonable basis for the identification of biomarkers and novel individualised treatments. Future clinical and laboratory trials would also focus on the diverse relationships between metabolism and ALS to address the issue of whether targeting deficient metabolism in ALS is an effective way to change disease progression. In this review, we focus on the detailed pathogenesis of ALS and highlight principal genes, i.e., SOD1, TDP-43, C9orf72, and FUS, targeted therapeutic approaches of ALS. An attempt is made to provide up-to-date information on clinical outcomes, including various biomarkers which are thought to be important players in early ALS detection.
Collapse
Affiliation(s)
- Vivek Chavda
- Department of Pharmaceutic, L M College of Pharmacy, Ahmedabad - 380009 (India)
| | - Chirag Patel
- Department of Pharmacology, L M College of Pharmacy, Ahmedabad - 380009 (India)
| | - Dharti Modh
- Department of pharmaceutical chemistry, Poona college of pharmacy, Bharti vidhyapith, Pune - 411030 (India)
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering at Erciyes University, Kayseri, Turkey
- ERNAM - Nanotechnology Research and Application Center, Erciyes University, Kayseri 38039, Turkey
| | - Shreya S Sonak
- Department of pharmaceutical chemistry, Poona college of pharmacy, Bharti vidhyapith, Pune - 411030 (India)
| | - Nafisa K Munshi
- Department of pharmaceutical chemistry, Poona college of pharmacy, Bharti vidhyapith, Pune - 411030 (India)
| | - Krishna Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences and National Health Laboratory Service, University of the Free State, Bloemfontein 9300, South Africa
| | - Arun Soni
- Department of Pharmacology, SSR College of Pharmacy, Silvassa, Dadra and Nagar Haveli - 396230(India)
| | - Sonal Pandey
- Research and Development, Meril Diagnostic Pvt. Ltd, Vapi - 396191 (India)
| |
Collapse
|
9
|
Gilmore M, Elman L, Babu S, Andres P, Floeter MK. Measuring disease progression in primary lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2021; 21:59-66. [PMID: 33602016 DOI: 10.1080/21678421.2020.1837179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Quantitative measures of disease severity are essential outcome measures for clinical trials. The slow progression of disease in primary lateral sclerosis (PLS) requires clinical measures that are sensitive to changes occurring within the time frame of a clinical trial. Proposed clinical outcome measures include the PLS functional rating scale (PLSFRS), burden scores derived from clinical examination findings, and quantitative measures of motor performance. The PLSFRS has good inter-rater reliability and showed greater longitudinal change over 6- and 12-months compared to the revised ALS functional rating scale. Examination-based upper motor neuron burden (UMNB) scales also have good reliability, and longitudinal studies are in process. Quantitative measures of strength, dexterity, gait, and speech have the potential to provide objective and precise measures of clinical change, but have been the least studied in persons with PLS.
Collapse
Affiliation(s)
- Madison Gilmore
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Lauren Elman
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Suma Babu
- Sean M Healy & AMG Center for ALS, Massachusetts General Hospital, Boston, MA, USA
| | - Patricia Andres
- Neurological Clinical Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Mary Kay Floeter
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| |
Collapse
|
10
|
Barohn RJ, Fink JK, Heiman-Patterson T, Huey ED, Murphy J, Statland JM, Turner MR, Elman L. The clinical spectrum of primary lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2021; 21:3-10. [PMID: 33602013 DOI: 10.1080/21678421.2020.1837178] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Primary lateral sclerosis is a distinct entity that has recently been classified as a "restricted phenotype" of ALS. It is characterized by a pattern of isolated upper motor neuron involvement that often begins in the legs and spreads diffusely. Distinction from other conditions requires careful consideration of clinical presentation and time course of disease. Mills' Syndrome is a rare unilateral variant of primary lateral sclerosis. Cognitive and behavioral involvement may occur.
Collapse
Affiliation(s)
- Richard J Barohn
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - John K Fink
- Department of Neurology, Ann Arbor Veterans Affairs Medical Center, University of Michigan, Ann Arbor, MI, USA
| | - Terry Heiman-Patterson
- Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Edward D Huey
- College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Jennifer Murphy
- Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Jeffrey M Statland
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Martin R Turner
- Nuffield Department of Neurosciences, University of Oxford, Oxford, UK
| | - Lauren Elman
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Lipidomics study of plasma from patients suggest that ALS and PLS are part of a continuum of motor neuron disorders. Sci Rep 2021; 11:13562. [PMID: 34193885 PMCID: PMC8245424 DOI: 10.1038/s41598-021-92112-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 12/14/2020] [Indexed: 12/04/2022] Open
Abstract
Motor neuron disorders (MND) include a group of pathologies that affect upper and/or lower motor neurons. Among them, amyotrophic lateral sclerosis (ALS) is characterized by progressive muscle weakness, with fatal outcomes only in a few years after diagnosis. On the other hand, primary lateral sclerosis (PLS), a more benign form of MND that only affects upper motor neurons, results in life-long progressive motor dysfunction. Although the outcomes are quite different, ALS and PLS present with similar symptoms at disease onset, to the degree that both disorders could be considered part of a continuum. These similarities and the lack of reliable biomarkers often result in delays in accurate diagnosis and/or treatment. In the nervous system, lipids exert a wide variety of functions, including roles in cell structure, synaptic transmission, and multiple metabolic processes. Thus, the study of the absolute and relative concentrations of a subset of lipids in human pathology can shed light into these cellular processes and unravel alterations in one or more pathways. In here, we report the lipid composition of longitudinal plasma samples from ALS and PLS patients initially, and after 2 years following enrollment in a clinical study. Our analysis revealed common aspects of these pathologies suggesting that, from the lipidomics point of view, PLS and ALS behave as part of a continuum of motor neuron disorders.
Collapse
|
12
|
Mitsumoto H, Garofalo DC, Gilmore M, Andrews L, Santella RM, Andrews H, McElhiney M, Murphy J, Nieves JW, Rabkin J, Hupf J, Horton DK, Mehta P, Factor-Litvak P. Case-control study in ALS using the National ALS Registry: lead and agricultural chemicals are potential risk factors. Amyotroph Lateral Scler Frontotemporal Degener 2021; 23:190-202. [PMID: 34137650 DOI: 10.1080/21678421.2021.1936556] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Objective: To identify occupational risk factors for ALS using well-characterized participants with ALS (P-ALS), sibling controls (S-controls), and matched population controls (P-controls) within the National ALS Registry. We also compared oxidative stress (OS) biomarkers between groups. Methods: P-ALS were recruited over 4 years. Demographic, socioeconomic, and medical data were ascertained from medical records and structured interviews. P-ALS were followed prospectively for 2 years or until death, whichever came sooner. S-controls and age-, sex-, race/ethnicity-, and residential location-matched P-controls were recruited over 3 years. Occupational exposure to lead and agricultural chemicals (ACs) were assigned by an occupational hygienist, blinded to case status. OS biomarkers in urine were measured. Results: P-ALS (mean age 62.8 years; 63% males) resided across the United States. Demographic and socioeconomic variables did not differ among P-ALS, S-controls, and P-controls. P-ALS were more likely to report occupations with exposure to lead (adjusted OR (aOR)=2.3, 95% CI 1.1, 4.6) and ACs (aOR = 2.4, 95% CI 1.2, 4.6) compared to pooled controls. Among those with occupations with exposure to both lead and ACs, aOR was 7.2 (95% CI 2.0, 26.1). Urinary 8-oxo-dG was significantly elevated among P-ALS (11.07 ± 5.42 ng/mL) compared to S-controls, P-controls, or pooled controls (pooled 7.43 ± 5.42 ng/mL; p < 0.0001) but was not associated with occupational exposure to either lead or ACs. Conclusions: Findings reveal increased risk of ALS diagnosis among those with occupational exposure to lead and ACs and increased OS biomarkers among cases compared to controls. OS may be an important pathogenic mechanism in ALS.
Collapse
Affiliation(s)
- Hiroshi Mitsumoto
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Diana C Garofalo
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Madison Gilmore
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Leslie Andrews
- Department of Environmental Health, Columbia University, New York, NY, USA
| | - Regina M Santella
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Howard Andrews
- Department of Biostatistics, Mailman School of Public Health, Columbia University and New York State Psychiatric Institute, New York, NY, USA
| | - Martin McElhiney
- Department of Clinical Psychology, Columbia University and New York State Psychiatric Institute, New York, NY, USA
| | - Jennifer Murphy
- Department of Neurology, University of California, San Francisco, CA, USA, and
| | - Jeri W Nieves
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Judith Rabkin
- Department of Clinical Psychology, Columbia University and New York State Psychiatric Institute, New York, NY, USA
| | - Jonathan Hupf
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - D Kevin Horton
- Centers for Disease Control and Prevention/Agency for Toxic Substance and Disease Registry (CDC/ATSDR), Atlanta, GA, USA
| | - Paul Mehta
- Centers for Disease Control and Prevention/Agency for Toxic Substance and Disease Registry (CDC/ATSDR), Atlanta, GA, USA
| | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
13
|
Fullam T, Statland J. Upper Motor Neuron Disorders: Primary Lateral Sclerosis, Upper Motor Neuron Dominant Amyotrophic Lateral Sclerosis, and Hereditary Spastic Paraplegia. Brain Sci 2021; 11:brainsci11050611. [PMID: 34064596 PMCID: PMC8151104 DOI: 10.3390/brainsci11050611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/02/2021] [Accepted: 05/10/2021] [Indexed: 11/16/2022] Open
Abstract
Following the exclusion of potentially reversible causes, the differential for those patients presenting with a predominant upper motor neuron syndrome includes primary lateral sclerosis (PLS), hereditary spastic paraplegia (HSP), or upper motor neuron dominant ALS (UMNdALS). Differentiation of these disorders in the early phases of disease remains challenging. While no single clinical or diagnostic tests is specific, there are several developing biomarkers and neuroimaging technologies which may help distinguish PLS from HSP and UMNdALS. Recent consensus diagnostic criteria and use of evolving technologies will allow more precise delineation of PLS from other upper motor neuron disorders and aid in the targeting of potentially disease-modifying therapeutics.
Collapse
|
14
|
Vucic S, Kiernan MC, Menon P, Huynh W, Rynders A, Ho KS, Glanzman R, Hotchkin MT. Study protocol of RESCUE-ALS: A Phase 2, randomised, double-blind, placebo-controlled study in early symptomatic amyotrophic lateral sclerosis patients to assess bioenergetic catalysis with CNM-A u8 as a mechanism to slow diseas e progression. BMJ Open 2021; 11:e041479. [PMID: 33431491 PMCID: PMC7802642 DOI: 10.1136/bmjopen-2020-041479] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is an adult-onset, progressive and universally fatal neurodegenerative disorder. In Europe, Australia and Canada, riluzole is the only approved therapeutic agent for the treatment of ALS, while in the USA, riluzole and edaravone have been approved by the Food and Drug Administration (FDA) . Neither riluzole nor edaravone treatment has resulted in substantial disease-modifying effects. There is, therefore, an urgent need for drugs that result in safe and effective treatment. Here, we present the design and rationale for the phase 2 RESCUE-ALS study, investigating the novel nanocatalytic drug, CNM-Au8, as a therapeutic intervention that enhances the metabolic and energetic capacity of motor neurones. CNM-Au8 is an aqueous suspension of clean-surfaced, faceted gold nanocrystals that have extraordinary catalytic capabilities, that enhance efficiencies of key metabolic reactions, while simultaneously reducing levels of reactive oxygen species. This trial utilises a novel design by employing motor unit number index (MUNIX), measured by electromyography, as a quantitative measure of lower motor neurone loss and as an early marker of ALS disease progression. METHODS AND ANALYSIS This is a multicentre, randomised, double-blind, parallel group, placebo-controlled study of the efficacy, safety, pharmacokinetics and pharmacodynamics of CNM-Au8 in ALS patients. Patients will be randomised 1:1 to either receive 30 mg of CNM-Au8 once daily or matching placebo over a 36-week double-blind treatment period. Efficacy will be assessed as the change in motor neurone loss as measured by electromyography (eg, MUNIX, the primary endpoint; and secondary endpoints including MScanFit, motor unit size index, Split Hand Index, Neurophysiology Index). Exploratory endpoints include standard clinical and quality of life assessments. ETHICS AND DISSEMINATION RESCUE-ALS was approved by the Western Sydney Local Health District Human Research Ethics Committee (Ethics Ref: 2019/ETH12107). Results of the study will be submitted for publication in a peer-reviewed journal. TRIAL REGISTRATION NUMBER NCT04098406.
Collapse
Affiliation(s)
- Steve Vucic
- Department of Neurology, Westmead Hospital and Western Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Parvathi Menon
- Department of Neurology, Westmead Hospital and Western Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - William Huynh
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Karen S Ho
- Clene Nanomedicine, Salt Lake City, Utah, USA
| | | | | |
Collapse
|
15
|
Rojas P, Ramírez AI, Fernández-Albarral JA, López-Cuenca I, Salobrar-García E, Cadena M, Elvira-Hurtado L, Salazar JJ, de Hoz R, Ramírez JM. Amyotrophic Lateral Sclerosis: A Neurodegenerative Motor Neuron Disease With Ocular Involvement. Front Neurosci 2020; 14:566858. [PMID: 33071739 PMCID: PMC7544921 DOI: 10.3389/fnins.2020.566858] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that causes degeneration of the lower and upper motor neurons and is the most prevalent motor neuron disease. This disease is characterized by muscle weakness, stiffness, and hyperreflexia. Patients survive for a short period from the onset of the disease. Most cases are sporadic, with only 10% of the cases being genetic. Many genes are now known to be involved in familial ALS cases, including some of the sporadic cases. It has also been observed that, in addition to genetic factors, there are numerous molecular mechanisms involved in these pathologies, such as excitotoxicity, mitochondrial disorders, alterations in axonal transport, oxidative stress, accumulation of misfolded proteins, and neuroinflammation. This pathology affects the motor neurons, the spinal cord, the cerebellum, and the brain, but recently, it has been shown that it also affects the visual system. This impact occurs not only at the level of the oculomotor system but also at the retinal level, which is why the retina is being proposed as a possible biomarker of this pathology. The current review discusses the main aspects mentioned above related to ALS, such as the main genes involved, the most important molecular mechanisms that affect this pathology, its ocular involvement, and the possible usefulness of the retina as a biomarker.
Collapse
Affiliation(s)
- Pilar Rojas
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain.,Hospital General Universitario Gregorio Marañón, Instituto Oftálmico de Madrid, Madrid, Spain
| | - Ana I Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain.,OFTARED, ISCIII, Madrid, Spain.,Departamento de Inmunología Oftalmología y ORL, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, Madrid, Spain
| | - José A Fernández-Albarral
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain
| | - Inés López-Cuenca
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain
| | - Elena Salobrar-García
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain.,OFTARED, ISCIII, Madrid, Spain.,Departamento de Inmunología Oftalmología y ORL, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Cadena
- Hospital General Universitario Gregorio Marañón, Instituto Oftálmico de Madrid, Madrid, Spain
| | - Lorena Elvira-Hurtado
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain
| | - Juan J Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain.,OFTARED, ISCIII, Madrid, Spain.,Departamento de Inmunología Oftalmología y ORL, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, Madrid, Spain
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain.,OFTARED, ISCIII, Madrid, Spain.,Departamento de Inmunología Oftalmología y ORL, Facultad de Óptica y Optometría, Universidad Complutense de Madrid, Madrid, Spain
| | - José M Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain.,OFTARED, ISCIII, Madrid, Spain.,Departamento de Inmunología Oftalmología y ORL, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
16
|
Beswick E, Park E, Wong C, Mehta AR, Dakin R, Chandran S, Newton J, Carson A, Abrahams S, Pal S. A systematic review of neuropsychiatric and cognitive assessments used in clinical trials for amyotrophic lateral sclerosis. J Neurol 2020; 268:4510-4521. [PMID: 32910255 PMCID: PMC8563523 DOI: 10.1007/s00415-020-10203-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 02/07/2023]
Abstract
Background Up to 50% of people with amyotrophic lateral sclerosis (ALS) experience cognitive dysfunction, whilst depression and anxiety are reported in up to 44% and 33%, respectively. These symptoms impact on quality of life, and are associated with a poorer prognosis. Historically, outcomes in clinical trials have focused on the effect of candidate drugs on physical functioning. Methods We reviewed the past 25 years of clinical trials of investigative medicinal products in people with ALS, since the licensing of riluzole, and extracted data on frequency and type of assessment for neuropsychiatric symptoms and cognitive impairment. Trial registry databases, including WHO International Trials Registry, European Clinical Trials Register, clinicaltrials.gov, and PubMed, were systematically searched for Phase II, III or IV trials registered, completed or published between 01/01/1994 and 31/10/2019. No language restrictions were applied. Outcome measures, exclusion criteria and assessment tool used were extracted. Results 216 trials, investigating 26,326 people with ALS, were reviewed. 35% assessed neuropsychiatric symptoms, and 22% assessed cognition, as Exclusion Criteria or Outcome Measures. 3% (n = 6) of trials assessed neuropsychiatric symptoms as a Secondary Outcome Measure, and 4% (n = 8) assessed cognition as Outcome Measures; only one trial included assessments for both cognition and neuropsychiatric symptoms as Outcome Measures. Three ALS-specific assessments were used in six trials. Conclusions Trials for people with ALS have neglected the importance of neuropsychiatric symptoms and cognitive impairment. Evaluation of these extra-motor features is essential to understanding the impact of candidate drugs on all symptoms of ALS. PROPSERO registration CRD42020175612. Electronic supplementary material The online version of this article (10.1007/s00415-020-10203-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emily Beswick
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland.,Anne Rowling Regenerative Neurology Clinic, 49 Little France Crescent, EH16 4SB, Edinburgh, UK.,Euan MacDonald Centre for MND Research, The University of Edinburgh, Edinburgh, Scotland
| | - Emily Park
- The School of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, Scotland
| | - Charis Wong
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland.,Anne Rowling Regenerative Neurology Clinic, 49 Little France Crescent, EH16 4SB, Edinburgh, UK.,Euan MacDonald Centre for MND Research, The University of Edinburgh, Edinburgh, Scotland
| | - Arpan R Mehta
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland.,Anne Rowling Regenerative Neurology Clinic, 49 Little France Crescent, EH16 4SB, Edinburgh, UK.,Euan MacDonald Centre for MND Research, The University of Edinburgh, Edinburgh, Scotland.,UK Dementia Research Institute, The University of Edinburgh, Edinburgh, Scotland
| | - Rachel Dakin
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland.,Anne Rowling Regenerative Neurology Clinic, 49 Little France Crescent, EH16 4SB, Edinburgh, UK
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland.,Anne Rowling Regenerative Neurology Clinic, 49 Little France Crescent, EH16 4SB, Edinburgh, UK.,Euan MacDonald Centre for MND Research, The University of Edinburgh, Edinburgh, Scotland.,UK Dementia Research Institute, The University of Edinburgh, Edinburgh, Scotland
| | - Judith Newton
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland.,Anne Rowling Regenerative Neurology Clinic, 49 Little France Crescent, EH16 4SB, Edinburgh, UK
| | - Alan Carson
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland
| | - Sharon Abrahams
- Euan MacDonald Centre for MND Research, The University of Edinburgh, Edinburgh, Scotland.,Human Cognitive Neurosciences, Psychology, School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, Scotland
| | - Suvankar Pal
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, Scotland. .,Anne Rowling Regenerative Neurology Clinic, 49 Little France Crescent, EH16 4SB, Edinburgh, UK. .,Euan MacDonald Centre for MND Research, The University of Edinburgh, Edinburgh, Scotland.
| |
Collapse
|
17
|
Mitsumoto H, Garofalo DC, Santella RM, Sorenson EJ, Oskarsson B, Fernandes JAM, Andrews H, Hupf J, Gilmore M, Heitzman D, Bedlack RS, Katz JS, Barohn RJ, Kasarskis EJ, Lomen-Hoerth C, Mozaffar T, Nations SP, Swenson AJ, Factor-Litvak P. Plasma creatinine and oxidative stress biomarkers in amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:263-272. [PMID: 32276554 DOI: 10.1080/21678421.2020.1746810] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Objective: To determine the associations between plasma creatinine (PCr), plasma uric acid (PUA), and urinary oxidative stress (OS) biomarkers with the ALSFRS-R at baseline and survival in a large epidemiological cohort study (ALS COSMOS) with a well-phenotyped patient population (N = 355).Methods: Fasting plasma and first void urine samples were obtained. PCr, PUA, urinary 8-oxo-deoxy guanosine (8-oxodG), and 15-F2t-isoprostane (IsoP) were analyzed at baseline, near the midpoint of follow-up, and at the final blood draw (before death or withdrawal from study). We estimated associations between these biomarkers and the ALSFRS-R at baseline and survival.Results: At baseline, PCr correlated with ALSFRS-R (Spearman r = 0.30), percent (%) FVC (r = 0.20), PUA (r = 0.37), and 8-oxodG (r = -0.13, all p < 0.05). Baseline PCr significantly predicted survival (adjusted hazard ratio 0.28, p < 0.001). Time to death from baseline was shortest for those in the lowest two PCr quartiles relative to the highest two quartiles. PCr and ALSFRS-R values were significantly correlated at all three time points (baseline: r = 0.29, midpoint: r = 0.23, final: r = 0.38, all p < 0.001). PCr and PUA significantly declined over time, whereas OS biomarkers significantly increased over time.Conclusions: To date, PCr predicted survival the best, compared to PUA, 8-oxodG, and IsoP. Although PCr represents the degree of muscle mass, it may also represent complex biochemical changes in ALS. Because the field has no reliable prognostic biomarkers, the importance of PCr warrants further investigation through clinical studies in ALS.
Collapse
Affiliation(s)
- Hiroshi Mitsumoto
- Department of Neurology, Eleanor and Lou Gehrig ALS Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Diana C Garofalo
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| | - Regina M Santella
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, New York, NY, USA
| | | | | | - J Americo M Fernandes
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard Andrews
- Data Coordinating Center (DCC), Mailman School of Public Health Biostatistics Department, Columbia University Irving Medical Center, New York State Psychiatric Institute & Department of Psychiatry, Columbia University
| | - Jonathan Hupf
- Department of Neurology, Eleanor and Lou Gehrig ALS Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Madison Gilmore
- Department of Neurology, Eleanor and Lou Gehrig ALS Center, Columbia University Irving Medical Center, New York, NY, USA
| | | | | | - Jonathan S Katz
- Forbes Norris ALS Center, California Pacific Medical Center, San Francisco, CA, USA
| | - Richard J Barohn
- Department of Neurology, University of Kansas, San Francisco, CA, USA
| | | | | | - Tahseen Mozaffar
- Department of Neurology, University of California, Irvine, CA, USA
| | - Sharon P Nations
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern, Dallas, TX, USA, and
| | | | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
18
|
Mitsumoto H, Chiuzan C, Gilmore M, Zhang Y, Simmons Z, Paganoni S, Kisanuki YY, Zinman L, Jawdat O, Sorenson E, Floeter MK, Pioro EP, Fernandes Filho JAM, Heitzman D, Fournier CN, Oskarsson B, Heiman‐Patterson T, Maragakis N, Joyce N, Hayat G, Nations S, Scelsa S, Walk D, Elman L, Hupf J, McHale B. Primary lateral sclerosis (PLS) functional rating scale: PLS‐specific clinimetric scale. Muscle Nerve 2019; 61:163-172. [DOI: 10.1002/mus.26765] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/07/2019] [Accepted: 11/19/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Hiroshi Mitsumoto
- Department of Neurology, Eleanor and Lou Gehrig ALS CenterColumbia University Irvine Medical Center New York New York
| | - Codruta Chiuzan
- Department of BiostatisticsMailman School of Medicine, Columbia University New York New York
| | - Madison Gilmore
- Department of Neurology, Eleanor and Lou Gehrig ALS CenterColumbia University Irvine Medical Center New York New York
| | - Yuan Zhang
- Department of BiostatisticsMailman School of Medicine, Columbia University New York New York
| | - Zachary Simmons
- Department of NeurologyPennsylvania State University Hershey Pennsylvania
| | - Sabrina Paganoni
- Sean M. Healey & AMG Center for ALS, Department of NeurologyMassachusetts General Hospital Boston Massachusetts
- Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital, Harvard Medical School Boston Massachusetts
| | | | - Lorne Zinman
- Department of NeurologyUniversity of Toronto, Sunnybrook Hospital Toronto Ontario Canada
| | - Omar Jawdat
- Department of NeurologyUniversity of Kansas Kansas City Kansas
| | - Eric Sorenson
- Department of NeurologyMayo Clinic, Minnesota Rochester Minnesota
| | - Mary Kay Floeter
- Clinical Unit, National Institute of Neurological Diseases and Stroke Bethesda Maryland
| | - Erik P. Pioro
- Department of NeurologyCleveland Clinic Cleveland Ohio
| | | | | | | | - Bjorn Oskarsson
- Department of NeurologyMayo Clinic Jacksonville Jacksonville Florida
| | | | | | - Nanette Joyce
- Department of Neurology University of California Davis Davis California
| | - Ghazala Hayat
- Department of NeurologySt Louis University St Louis Missouri
| | - Sharon Nations
- Department of NeurologyUniversity of Texas Southwestern Dallas Texas
| | - Stephen Scelsa
- Department of NeurologyMount Sinai/Beth Israel Hospital New York New York
| | - David Walk
- Department of NeurologyUniversity of Minnesota Minneapolis Minnesota
| | - Lauren Elman
- Department of NeurologyUniversity of Pennsylvania Philadelphia Pennsylvania
| | - Jonathan Hupf
- Department of Neurology, Eleanor and Lou Gehrig ALS CenterColumbia University Irvine Medical Center New York New York
| | - Brittany McHale
- Department of Neurology, Eleanor and Lou Gehrig ALS CenterColumbia University Irvine Medical Center New York New York
| | | |
Collapse
|
19
|
Moreno-Martinez L, Calvo AC, Muñoz MJ, Osta R. Are Circulating Cytokines Reliable Biomarkers for Amyotrophic Lateral Sclerosis? Int J Mol Sci 2019; 20:ijms20112759. [PMID: 31195629 PMCID: PMC6600567 DOI: 10.3390/ijms20112759] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that has no effective treatment. The lack of any specific biomarker that can help in the diagnosis or prognosis of ALS has made the identification of biomarkers an urgent challenge. Multiple panels have shown alterations in levels of numerous cytokines in ALS, supporting the contribution of neuroinflammation to the progressive motor neuron loss. However, none of them is fully sensitive and specific enough to become a universal biomarker for ALS. This review gathers the numerous circulating cytokines that have been found dysregulated in both ALS animal models and patients. Particularly, it highlights the opposing results found in the literature to date, and points out another potential application of inflammatory cytokines as therapeutic targets.
Collapse
Affiliation(s)
- Laura Moreno-Martinez
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary-IIS Aragón, IA2-CITA, CIBERNED, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain.
| | - Ana Cristina Calvo
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary-IIS Aragón, IA2-CITA, CIBERNED, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain.
| | - María Jesús Muñoz
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary-IIS Aragón, IA2-CITA, CIBERNED, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain.
| | - Rosario Osta
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary-IIS Aragón, IA2-CITA, CIBERNED, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain.
| |
Collapse
|
20
|
Abstract
Although we currently have two, approved, disease-modifying drugs for the treatment of amyotrophic lateral sclerosis (ALS), we are in disperate need for more efficacious treatment. To aggressively test for newer therapies, we must develop reliable objective biomarkers to supplement clinical outcome measures. Many biomarker candidates have been actively and vigorously investigated. Among neurophysiological biomarkers, transcranial magnetic stimulation (TMS)-based biomarkers show potential in exploring disease mechanisms. Neuroimaging biomarkers have high specificity in diagnosing ALS but are an expensive endeavor and are not sensitive enough to detect changes over time of the disease. Among fluid-based biochemical biomarkers, creatinine (Crn) and uric acids (UA), which have been known for decades, may prove to be highly promising biomarkers that can predict disease progression. They can be easily tested in any clinical trials because the costs are minimal. Although known for some time, neurofilaments (NF), either phosphorylated-NF heavy subunit (pNFH) or NF light subunit (NFL), have emerged as "new" biomarkers using specific antibodies. They appear to be highly specific and sensitive in diagnosing ALS, yet they may be insensitive to assess changes in disease over time. These two NF biomarkers along with Crn and UA should be explored extensively in future clinical trials and any other clinical studies in ALS. Yet, we still need newer, more innovative, and reliable biomarkers for future ALS research. Fortunatley, aggressive investigations appear to be currently underway.
Collapse
Affiliation(s)
- Hiroshi Mitsumoto
- Wesley J Howe Professor of Neurology (at CUMC), Eleanor and Lou Gehrig ALS Center, Department of Neurology, Columbia University Medical Center (CUMC)
| | | |
Collapse
|
21
|
Longitudinal Screening Detects Cognitive Stability and Behavioral Deterioration in ALS Patients. Behav Neurol 2018; 2018:5969137. [PMID: 30515252 PMCID: PMC6234441 DOI: 10.1155/2018/5969137] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 08/08/2018] [Accepted: 09/10/2018] [Indexed: 12/03/2022] Open
Abstract
Objective To evaluate longitudinal cognitive/behavioral change over 12 months in participants enrolled in the ALS Multicenter Cohort Study of Oxidative Stress (ALS COSMOS). Methods We analyzed data from 294 ALS participants, 134 of whom were studied serially. Change over time was evaluated controlling for age, sex, symptom duration, education, race, and ethnicity. Using multiple regression, we evaluated associations among decline in ALS Functional Rating Scale-Revised (ALSFRS-R) scores, forced vital capacity (FVC), and cognitive/behavioral changes. Change in cognitive/behavioral subgroups was assessed using one-way analyses of covariance. Results Participants with follow-up data had fewer baseline behavior problems compared to patients without follow-up data. We found significant worsening of behavior (ALS Cognitive Behavioral Screen (ALS CBS) behavioral scale, p < 0.001; Frontal Behavioral Inventory-ALS (FBI-ALS) disinhibition subscale, p = 0.044). Item analysis suggested change in frustration tolerance, insight, mental rigidity, and interests (p < 0.05). Changes in ALSFRS-R correlated with the ALS CBS. Worsening disinhibition (FBI-ALS) did not correlate with ALSFRS-R, FVC, or disease duration. Conclusion We did not detect cognitive change. Behavioral change was detected, and increased disinhibition was found among patients with abnormal baseline behavioral scores. Disinhibition changes did not correlate with disease duration or progression. Baseline behavioral problems were associated with advanced, rapidly progressive disease and study attrition.
Collapse
|
22
|
Khairoalsindi OA, Abuzinadah AR. Maximizing the Survival of Amyotrophic Lateral Sclerosis Patients: Current Perspectives. Neurol Res Int 2018; 2018:6534150. [PMID: 30159171 PMCID: PMC6109498 DOI: 10.1155/2018/6534150] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 07/26/2018] [Indexed: 01/08/2023] Open
Abstract
Amyotrophic lateral sclerosis is a neurodegenerative disease that leads to loss of the upper and lower motor neurons. Almost 90% of all cases occur in the sporadic form, with the rest occurring in the familial form. The disease has a poor prognosis, with only two disease-modifying drugs approved by the United States Food and Drug Administration (FDA). The approved drugs for the disease have very limited survival benefits. Edaravone is a new FDA-approved medication that may slow the disease progression by 33% in a selected subgroup of ALS patients. This paper covers the various interventions that may provide survival benefits, such as early diagnosis, medications, gene therapy, stem cell therapy, diet, nutritional supplements, multidisciplinary clinics, and mechanical invasive and noninvasive ventilation. The recent data on masitinib, the role of enteral feeding, gene therapy, and stem cell therapy is discussed.
Collapse
Affiliation(s)
| | - Ahmad R. Abuzinadah
- King Abdulaziz University, Internal Medicine Department, Neurology Division, Jeddah, Saudi Arabia
| |
Collapse
|
23
|
Nieves JW, Gennings C, Factor-Litvak P, Hupf J, Singleton J, Sharf V, Oskarsson B, Fernandes Filho JAM, Sorenson EJ, D'Amico E, Goetz R, Mitsumoto H. Association Between Dietary Intake and Function in Amyotrophic Lateral Sclerosis. JAMA Neurol 2017; 73:1425-1432. [PMID: 27775751 DOI: 10.1001/jamaneurol.2016.3401] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Importance There is growing interest in the role of nutrition in the pathogenesis and progression of amyotrophic lateral sclerosis (ALS). Objective To evaluate the associations between nutrients, individually and in groups, and ALS function and respiratory function at diagnosis. Design, Setting, and Participants A cross-sectional baseline analysis of the Amyotrophic Lateral Sclerosis Multicenter Cohort Study of Oxidative Stress study was conducted from March 14, 2008, to February 27, 2013, at 16 ALS clinics throughout the United States among 302 patients with ALS symptom duration of 18 months or less. Exposures Nutrient intake, measured using a modified Block Food Frequency Questionnaire (FFQ). Main Outcomes and Measures Amyotrophic lateral sclerosis function, measured using the ALS Functional Rating Scale-Revised (ALSFRS-R), and respiratory function, measured using percentage of predicted forced vital capacity (FVC). Results Baseline data were available on 302 patients with ALS (median age, 63.2 years [interquartile range, 55.5-68.0 years]; 178 men and 124 women). Regression analysis of nutrients found that higher intakes of antioxidants and carotenes from vegetables were associated with higher ALSFRS-R scores or percentage FVC. Empirically weighted indices using the weighted quantile sum regression method of "good" micronutrients and "good" food groups were positively associated with ALSFRS-R scores (β [SE], 2.7 [0.69] and 2.9 [0.9], respectively) and percentage FVC (β [SE], 12.1 [2.8] and 11.5 [3.4], respectively) (all P < .001). Positive and significant associations with ALSFRS-R scores (β [SE], 1.5 [0.61]; P = .02) and percentage FVC (β [SE], 5.2 [2.2]; P = .02) for selected vitamins were found in exploratory analyses. Conclusions and Relevance Antioxidants, carotenes, fruits, and vegetables were associated with higher ALS function at baseline by regression of nutrient indices and weighted quantile sum regression analysis. We also demonstrated the usefulness of the weighted quantile sum regression method in the evaluation of diet. Those responsible for nutritional care of the patient with ALS should consider promoting fruit and vegetable intake since they are high in antioxidants and carotenes.
Collapse
Affiliation(s)
- Jeri W Nieves
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York2Clinical Research Center, Helen Hayes Hospital, West Haverstraw, New York
| | - Chris Gennings
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Jonathan Hupf
- Department of Neurology, Columbia University, New York, New York
| | | | - Valerie Sharf
- Department of Neurology, Columbia University, New York, New York
| | - Björn Oskarsson
- Department of Neurology, University of California-Davis, Sacramento
| | | | | | | | - Ray Goetz
- Department of Psychiatry, New York State Psychiatric Institute, New York
| | | | | |
Collapse
|
24
|
Abstract
AbstractBackground: Pathophysiological mechanisms that contribute to neurodegeneration in Amyotrophic Lateral Sclerosis (ALS) include oxidative stress and inflammation. We conducted a preliminary study to explore these mechanisms, to discuss their link in ALS, and to determine the feasibility of incorporating this combined analysis into current biomarkers research. Methods: We enrolled 10 ALS patients and 10 controls. We measured the activities of glutathione peroxidase, glutathione reductase, superoxyde dismutase (SOD), and the levels of serum total antioxidant status (TAS), malondialdehyde (MDA), 8-hydroxy-2’-deoxyguanosine (8-OHdG), and glutathione status (e.g. glutathione disulfide, GSSG/reduced glutathione, GSH). We analysed the concentrations of homocysteine, several cytokines, vitamins and metals by standard methods used in routine practice. Results: There was a significant decrease in TAS levels (p=0.027) and increase in 8-OHdG (p=0.014) and MDA (p=0.011) levels in ALS patients. We also observed a significantly higher GSSG/GSH ratio (p=0.022), and IL-6 (p=0.0079) and IL-8 (p=0.009) concentrations in ALS patients. Correlations were found between biological and clinical markers (homosysteine vs. clinical status at diagnosis, p=0.02) and between some biological markers such as IL-6 vs. GSSG/GSH (p=0.045) or SOD activity (p=0.017). Conclusion: We confirmed the systemic alteration of both the redox and the inflammation status in ALS patients, and we observed a link with some clinical parameters. These promising results encourage us to pursue this study with collection of combined oxidative stress and inflammatory markers.
Collapse
|
25
|
Rabkin J, Goetz R, Murphy JM, Factor-Litvak P, Mitsumoto H. Cognitive impairment, behavioral impairment, depression, and wish to die in an ALS cohort. Neurology 2016; 87:1320-8. [PMID: 27496520 PMCID: PMC5573192 DOI: 10.1212/wnl.0000000000003035] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/17/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To evaluate relationships among cognitive, behavioral, and psychiatric/psychosocial measures assessed in a multicenter cohort of patients with amyotrophic lateral sclerosis (ALS). METHODS Recently diagnosed patients with definite or probable ALS diagnosis were administered 7 standardized psychiatric/psychosocial measures, including the Patient Health Questionnaire for diagnosis of depression and elicitation of wish to die. The Cognitive Behavioral Screen was used to classify both cognitive and behavioral impairment (emotional-interpersonal function). An ALS version of the Frontal Behavioral Inventory and Mini-Mental State Examination were also administered. RESULTS Of 247 patients included, 79 patients (32%) had neither cognitive nor behavioral impairment, 100 (40%) had cognitive impairment, 23 (9%) had behavioral impairment, and 45 (18%) had comorbid cognitive and behavioral decline. Cognitive impairment, when present, was in the mild range for 90% and severe for 10%. Thirty-one patients (12%) had a major or minor depressive disorder (DSM-IV criteria). Cognitive impairment was unrelated to all psychiatric/psychosocial measures. In contrast, patients with behavioral impairment reported more depressive symptoms, greater hopelessness, negative mood, and more negative feedback from spouse or caregiver. A wish to die was unrelated to either cognitive or behavioral impairment. CONCLUSIONS While we found no association between cognitive impairment and depression or any measure of distress, behavioral impairment was strongly associated with depressive symptoms and diagnoses although seldom addressed by clinicians. Thoughts about ending life were unrelated to either cognitive or behavioral changes, a finding useful to consider in the context of policy debate about physician-assisted death.
Collapse
Affiliation(s)
- Judith Rabkin
- From the New York State Psychiatric Institute (J.R., R.G.), Department of Psychiatry (J.R., R.G.), and Eleanor and Lou Gehrig MDA/ALS Research Center, Department of Neurology (H.M.), Columbia University Medical Center, New York, NY; Department of Neurology (J.M.M.), University of California San Francisco; and Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University, New York, NY.
| | - Raymond Goetz
- From the New York State Psychiatric Institute (J.R., R.G.), Department of Psychiatry (J.R., R.G.), and Eleanor and Lou Gehrig MDA/ALS Research Center, Department of Neurology (H.M.), Columbia University Medical Center, New York, NY; Department of Neurology (J.M.M.), University of California San Francisco; and Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University, New York, NY
| | - Jennifer Mary Murphy
- From the New York State Psychiatric Institute (J.R., R.G.), Department of Psychiatry (J.R., R.G.), and Eleanor and Lou Gehrig MDA/ALS Research Center, Department of Neurology (H.M.), Columbia University Medical Center, New York, NY; Department of Neurology (J.M.M.), University of California San Francisco; and Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University, New York, NY
| | - Pam Factor-Litvak
- From the New York State Psychiatric Institute (J.R., R.G.), Department of Psychiatry (J.R., R.G.), and Eleanor and Lou Gehrig MDA/ALS Research Center, Department of Neurology (H.M.), Columbia University Medical Center, New York, NY; Department of Neurology (J.M.M.), University of California San Francisco; and Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University, New York, NY
| | | |
Collapse
|
26
|
Abstract
The causes of amyotrophic lateral sclerosis (ALS) are largely unknown, and may always be multiple, including environmental factors. Monogenetic determinants of ALS are involved in roughly 20% of all cases (including 10% familial cases). Less well understood multigenetic causes may contribute to another 20% to 80%. Environmental factors likely play a role in the development of ALS in susceptible individuals, but proved causation remains elusive. This article discusses the possible factors of male gender (males are selectively exposed to different influences, or genetically predisposed to be susceptible), smoking, military service, exercise, electrical exposure, heavy metals, agricultural chemicals, and geographic clusters.
Collapse
Affiliation(s)
- Björn Oskarsson
- UC Davis Multidisciplinary ALS Clinic, An ALS Association Certified Center of Excellence, University of California Davis Medical Center, 4860 Y Street, Suite 3700, Sacramento, CA 95817, USA.
| | - D Kevin Horton
- Division of Toxicology and Human Health Sciences, ATSDR/CDC, 4770 Buford Highway Northeast, Atlanta, GA 30341, USA
| | - Hiroshi Mitsumoto
- The Eleanor and Lou Gehrig MDA/ALS Research Center, The Neurological Institute, Columbia University Medical Center, 710 West 168th Street, Floor 9, New York, NY 10032, USA
| |
Collapse
|
27
|
Murphy J, Factor-Litvak P, Goetz R, Lomen-Hoerth C, Nagy PL, Hupf J, Singleton J, Woolley S, Andrews H, Heitzman D, Bedlack RS, Katz JS, Barohn RJ, Sorenson EJ, Oskarsson B, Fernandes Filho JAM, Kasarskis EJ, Mozaffar T, Rollins YD, Nations SP, Swenson AJ, Koczon-Jaremko BA, Mitsumoto H. Cognitive-behavioral screening reveals prevalent impairment in a large multicenter ALS cohort. Neurology 2016; 86:813-20. [PMID: 26802094 PMCID: PMC4793785 DOI: 10.1212/wnl.0000000000002305] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 11/05/2015] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES To characterize the prevalence of cognitive and behavioral symptoms using a cognitive/behavioral screening battery in a large prospective multicenter study of amyotrophic lateral sclerosis (ALS). METHODS Two hundred seventy-four patients with ALS completed 2 validated cognitive screening tests and 2 validated behavioral interviews with accompanying caregivers. We examined the associations between cognitive and behavioral performance, demographic and clinical data, and C9orf72 mutation data. RESULTS Based on the ALS Cognitive Behavioral Screen cognitive score, 6.5% of the sample scored below the cutoff score for frontotemporal lobar dementia, 54.2% scored in a range consistent with ALS with mild cognitive impairment, and 39.2% scored in the normal range. The ALS Cognitive Behavioral Screen behavioral subscale identified 16.5% of the sample scoring below the dementia cutoff score, with an additional 14.1% scoring in the ALS behavioral impairment range, and 69.4% scoring in the normal range. CONCLUSIONS This investigation revealed high levels of cognitive and behavioral impairment in patients with ALS within 18 months of symptom onset, comparable to prior investigations. This investigation illustrates the successful use and scientific value of adding a cognitive-behavioral screening tool in studies of motor neuron diseases, to provide neurologists with an efficient method to measure these common deficits and to understand how they relate to key clinical variables, when extensive neuropsychological examinations are unavailable. These tools, developed specifically for patients with motor impairment, may be particularly useful in patient populations with multiple sclerosis and Parkinson disease, who are known to have comorbid cognitive decline.
Collapse
Affiliation(s)
- Jennifer Murphy
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT.
| | - Pam Factor-Litvak
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Raymond Goetz
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Catherine Lomen-Hoerth
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Peter L Nagy
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Jonathan Hupf
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Jessica Singleton
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Susan Woolley
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Howard Andrews
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Daragh Heitzman
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Richard S Bedlack
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Jonathan S Katz
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Richard J Barohn
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Eric J Sorenson
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Björn Oskarsson
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - J Americo M Fernandes Filho
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Edward J Kasarskis
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Tahseen Mozaffar
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Yvonne D Rollins
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Sharon P Nations
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Andrea J Swenson
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | - Boguslawa A Koczon-Jaremko
- From the Department of Neurology (J.M., C.L.-H.), UCSF; Department of Epidemiology (P.F.-L.), Mailman School of Public Health, Columbia University Medical Center (CUMC); Department of Psychiatry (R.G.), New York State Psychiatric Institute and CUMC; Department of Pathology and Cell Biology (P.L.N), Columbia University; Eleanor and Lou Gehrig MDA/ALS Research Center (J.H., J.S., H.M.), Department of Neurology, CUMC, New York, NY; California Pacific Medical Center (S.W., J.S.K.), San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A.), Mailman School of Medicine, CUMC, New York, NY; Texas Neurology (D.H.), P.A., Dallas, TX; Duke University (R.S.B.), Durham, NC; Department of Neurology (R.J.B.), University of Kansas; Mayo Clinic (E.J.S.), Rochester, MN; University of California, Davis (B.O.), Sacramento, CA; University of Nebraska Medical Center (J.A.M.F.F.), Omaha, NE; University of Kentucky (E.J.K.), Lexington, KY; University of California, Irvine (T.M.), Orange, CA; University of Colorado, Denver (Y.D.R.), Aurora, CO; University of Texas-Southwestern (S.P.N.), Dallas, TX; University of Iowa (A.J.S.), Iowa City, IA; and Hospital for Special Care (B.A.K.-J.), New Britain, CT
| | | |
Collapse
|
28
|
Zarei S, Carr K, Reiley L, Diaz K, Guerra O, Altamirano PF, Pagani W, Lodin D, Orozco G, Chinea A. A comprehensive review of amyotrophic lateral sclerosis. Surg Neurol Int 2015; 6:171. [PMID: 26629397 PMCID: PMC4653353 DOI: 10.4103/2152-7806.169561] [Citation(s) in RCA: 400] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/12/2015] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a late-onset fatal neurodegenerative disease affecting motor neurons with an incidence of about 1/100,000. Most ALS cases are sporadic, but 5–10% of the cases are familial ALS. Both sporadic and familial ALS (FALS) are associated with degeneration of cortical and spinal motor neurons. The etiology of ALS remains unknown. However, mutations of superoxide dismutase 1 have been known as the most common cause of FALS. In this study, we provide a comprehensive review of ALS. We cover all aspects of the disease including epidemiology, comorbidities, environmental risk factor, molecular mechanism, genetic factors, symptoms, diagnostic, treatment, and even the available supplement and management of ALS. This will provide the reader with an advantage of receiving a broad range of information about the disease.
Collapse
Affiliation(s)
- Sara Zarei
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Karen Carr
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Luz Reiley
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Kelvin Diaz
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Orleiquis Guerra
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | | | - Wilfredo Pagani
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Daud Lodin
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Gloria Orozco
- Department of Medicine, San Juan Bautista School of Medicine, Caguas, USA
| | - Angel Chinea
- Neurologist, Caribbean Neurological Center, Caguas, USA
| |
Collapse
|
29
|
Wagner L, Rechtman L, Jordan H, Ritsick M, Sanchez M, Sorenson E, Kaye W. State and metropolitan area-based amyotrophic lateral sclerosis (ALS) surveillance. Amyotroph Lateral Scler Frontotemporal Degener 2015; 17:128-34. [PMID: 26399278 PMCID: PMC4732418 DOI: 10.3109/21678421.2015.1074699] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Our objective was to develop state and metropolitan area-based surveillance projects to describe the characteristics of those with ALS and to assist with evaluating the completeness of the National ALS Registry. Because the literature suggested that ethnic/racial minorities have lower incidence of ALS, three state and eight metropolitan areas were selected to over-represent ethnic/racial minorities to have a sufficient number of minority patients. Project activities relied on reports from medical providers and medical records abstraction. The project areas represented approximately 27% of the U.S. population. The combined racial and ethnic distribution of these areas is 64.4% white, 16.0% African-American, 6.7% Asian, and 28.3% Hispanic. Most neurologists did not diagnose or provide care for ALS patients. The number of unique patients reported was close to expected (5883 vs. 6673). Age and gender distribution of patients was similar to the literature. The crude average annual incidence rate was 1.52 per 100,000 person-years, CI 1.44–1.61, and the 2009 prevalence rate was 3.84 per 100,000 population, CI 3.70–3.97. In conclusion, this study represents the largest number of clinically diagnosed ALS patients reported by neurologists in the U.S. Comparison of these data with those in the National ALS Registry will help evaluate the completeness of administrative databases.
Collapse
Affiliation(s)
- Laurie Wagner
- a McKing Consulting Corporation , Atlanta, Georgia , USA
| | | | - Heather Jordan
- a McKing Consulting Corporation , Atlanta, Georgia , USA
| | - Maggie Ritsick
- a McKing Consulting Corporation , Atlanta, Georgia , USA
| | - Marchelle Sanchez
- b Agency for Toxic Substances and Disease Registry , Atlanta, Georgia , USA
| | | | - Wendy Kaye
- a McKing Consulting Corporation , Atlanta, Georgia , USA
| |
Collapse
|
30
|
Henry KA, Fagliano J, Jordan HM, Rechtman L, Kaye WE. Geographic Variation of Amyotrophic Lateral Sclerosis Incidence in New Jersey, 2009-2011. Am J Epidemiol 2015; 182:512-9. [PMID: 26041711 PMCID: PMC4564938 DOI: 10.1093/aje/kwv095] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/03/2015] [Indexed: 01/03/2023] Open
Abstract
Few analyses in the United States have examined geographic variation and socioeconomic disparities in amyotrophic lateral sclerosis (ALS) incidence, because of lack of population-based incidence data. In this analysis, we used population-based ALS data to identify whether ALS incidence clusters geographically and to determine whether ALS risk varies by area-based socioeconomic status (SES). This study included 493 incident ALS cases diagnosed (via El Escorial criteria) in New Jersey between 2009 and 2011. Geographic variation and clustering of ALS incidence was assessed using a spatial scan statistic and Bayesian geoadditive models. Poisson regression was used to estimate the associations between ALS risk and SES based on census-tract median income while controlling for age, sex, and race. ALS incidence varied across and within counties, but there were no statistically significant geographic clusters. SES was associated with ALS incidence. After adjustment for age, sex, and race, the relative risk of ALS was significantly higher (relative risk (RR) = 1.37, 95% confidence interval (CI): 1.02, 1.82) in the highest income quartile than in the lowest. The relative risk of ALS was significantly lower among blacks (RR = 0.57, 95% CI: 0.39, 0.83) and Asians (RR = 0.63, 95% CI: 0.41, 0.97) than among whites. Our findings suggest that ALS incidence in New Jersey appears to be associated with SES and race.
Collapse
Affiliation(s)
- Kevin A. Henry
- Correspondence to Dr. Kevin A. Henry, Department of Geography and Urban Studies, College of Liberal Arts, Temple University, Gladfelter Hall, Room 313b, Philadelphia, PA 19122 (e-mail: )
| | | | | | | | | |
Collapse
|
31
|
Zuo L, Zhou T, Pannell BK, Ziegler AC, Best TM. Biological and physiological role of reactive oxygen species--the good, the bad and the ugly. Acta Physiol (Oxf) 2015; 214:329-48. [PMID: 25912260 DOI: 10.1111/apha.12515] [Citation(s) in RCA: 290] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/27/2015] [Accepted: 04/21/2015] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS) are chemically reactive molecules that are naturally produced within biological systems. Research has focused extensively on revealing the multi-faceted and complex roles that ROS play in living tissues. In regard to the good side of ROS, this article explores the effects of ROS on signalling, immune response and other physiological responses. To review the potentially bad side of ROS, we explain the consequences of high concentrations of molecules that lead to the disruption of redox homeostasis, which induces oxidative stress damaging intracellular components. The ugly effects of ROS can be observed in devastating cardiac, pulmonary, neurodegenerative and other disorders. Furthermore, this article covers the regulatory enzymes that mitigate the effects of ROS. Glutathione peroxidase, superoxide dismutase and catalase are discussed in particular detail. The current understanding of ROS is incomplete, and it is imperative that future research be performed to understand the implications of ROS in various therapeutic interventions.
Collapse
Affiliation(s)
- L. Zuo
- Radiologic Sciences and Respiratory Therapy Division; School of Health and Rehabilitation Sciences; The Ohio State University College of Medicine; Columbus OH USA
- Biophysics Graduate Program; The Ohio State University; Columbus OH USA
| | - T. Zhou
- Radiologic Sciences and Respiratory Therapy Division; School of Health and Rehabilitation Sciences; The Ohio State University College of Medicine; Columbus OH USA
- Biophysics Graduate Program; The Ohio State University; Columbus OH USA
| | - B. K. Pannell
- Radiologic Sciences and Respiratory Therapy Division; School of Health and Rehabilitation Sciences; The Ohio State University College of Medicine; Columbus OH USA
| | - A. C. Ziegler
- Radiologic Sciences and Respiratory Therapy Division; School of Health and Rehabilitation Sciences; The Ohio State University College of Medicine; Columbus OH USA
| | - T. M. Best
- Division of Sports Medicine; Department of Family Medicine; Sports Health & Performance Institute; The Ohio State University Wexner Medical Center; Columbus OH USA
| |
Collapse
|
32
|
Rabkin JG, Goetz R, Factor-Litvak P, Hupf J, McElhiney M, Singleton J, Mitsumoto H. Depression and wish to die in a multicenter cohort of ALS patients. Amyotroph Lateral Scler Frontotemporal Degener 2015; 16:265-73. [PMID: 25482273 PMCID: PMC4441849 DOI: 10.3109/21678421.2014.980428] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Our objective was to determine prevalence of depressive disorders and wish to die at the baseline visit of a longitudinal multisite study of patients with ALS. Structured telephone interviews were conducted with patients diagnosed in past 18 months at 16 U.S. ALS centers. Demographic, medical, psychiatric and other psychological measures were administered. Of 329 patients assessed, mean ALSFRS-R score was 36.6; 88% (289/329) had no depressive disorder, 7% (24/329) had minor depression, and 5% (16/329) had current major depressive disorder (DSM-IV criteria). Demographic, financial and employment factors were unrelated to depression, as were duration of ALS symptoms and respiratory status, although depressed patients had lower scores on the total ALSFRS-R (p = 0.004) and gross motor function (p < 0.001). Depressed patients reported less pleasure, greater suffering, weariness and anxiety, more stress, were less hopeful, felt less control over illness management, reported lower quality of life, more often had thoughts about ending their lives and hastening death (all p < 0.001). Of the 62 patients (19% of the sample) who expressed a wish to die, only 37% (23/62) were clinically depressed. In conclusion, depressive disorders are not necessarily to be expected of ALS patients. Wish to die is not always expressed in the context of depression and does not necessarily represent psychopathology as such.
Collapse
Affiliation(s)
- Judith G Rabkin
- New York State Psychiatric Institute and Department of Psychiatry, Columbia University College of Physicians and Surgeons , New York
| | | | | | | | | | | | | |
Collapse
|
33
|
Mitsumoto H, Nagy PL, Gennings C, Murphy J, Andrews H, Goetz R, Floeter MK, Hupf J, Singleton J, Barohn RJ, Nations S, Shoesmith C, Kasarskis E, Factor-Litvak P. Phenotypic and molecular analyses of primary lateral sclerosis. NEUROLOGY-GENETICS 2015; 1:e3. [PMID: 27066542 PMCID: PMC4821084 DOI: 10.1212/01.nxg.0000464294.88607.dd] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 03/17/2015] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To understand phenotypic and molecular characteristics of patients with clinically "definite" primary lateral sclerosis (PLS) in a prospective study. METHODS Six sites enrolled 41 patients who had pure upper motor neuron dysfunction, bulbar symptoms, a normal EMG done within 12 months of enrollment, and onset of symptoms ≥5 years before enrollment. For phenotypic analyses, 27 demographic, clinical, and cognitive variables were analyzed using the k-means clustering method. For molecular studies, 34 available DNA samples were tested for the C9ORF72 mutation, and exome sequencing was performed to exclude other neurologic diseases with known genetic cause. RESULTS K-means clustering using the 25 patients with complete datasets suggested that patients with PLS can be classified into 2 groups based on clinical variables, namely dysphagia, objective bulbar signs, and urinary urgency. Secondary analyses performed in all 41 patients and including only variables with complete data corroborated the results from the primary analysis. We found no evidence that neurocognitive variables are important in classifying patients with PLS. Molecular studies identified C9ORF72 expansion in one patient. Well-characterized pathogenic mutations were identified in SPG7, DCTN1, and PARK2. Most cases showed no known relevant mutations. CONCLUSIONS Cluster analyses based on clinical variables indicated at least 2 subgroups of clinically definite PLS. Molecular analyses further identified 4 cases with mutations associated with amyotrophic lateral sclerosis, Parkinson disease, and possibly hereditary spastic paraplegia. Phenotypic and molecular characterization is the first step in investigating biological clues toward the definition of PLS. Further studies with larger numbers of patients are essential.
Collapse
Affiliation(s)
- Hiroshi Mitsumoto
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Peter L Nagy
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Chris Gennings
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Jennifer Murphy
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Howard Andrews
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Raymond Goetz
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Mary Kay Floeter
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Jonathan Hupf
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Jessica Singleton
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Richard J Barohn
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Sharon Nations
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Christen Shoesmith
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Edward Kasarskis
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| | - Pam Factor-Litvak
- Department of Neurology (H.M., J.H., J.S.), Eleanor and Lou Gehrig MDA/ALS Research Center, Columbia University Medical Center (CUMC), New York, NY; Department of Pathology and Cell Biology (P.L.N.), Personalized Genomic Medicine Laboratory, CUMC, New York, NY; Department of Biostatistics (C.G.), Virginia Commonwealth University, Richmond, VA; Department of Neurology (J.M.), University of California, San Francisco, CA; Departments of Biostatistics and Psychiatry (H.A., R.G.), Mailman School of Medicine, CUMC, New York, NY; Clinical Neuroscience Program (M.K.F.), NINDS, NIH, Bethesda, MD; Department of Neurology (R.J.B.), University of Kansas, Lawrence, KS; Department of Neurology (S.N.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Neurology (C.S.), Western University, London, Ontario, Canada; Department of Neurology (E.K.), University of Kentucky, Lexington, KY; and Department of Epidemiology (P.F.-L), Mailman School of Public Health, CUMC, New York, NY
| |
Collapse
|
34
|
Jordan H, Fagliano J, Rechtman L, Lefkowitz D, Kaye W. Population-based surveillance of amyotrophic lateral sclerosis in New Jersey, 2009-2011. Neuroepidemiology 2014; 43:49-56. [PMID: 25323440 DOI: 10.1159/000365850] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 06/06/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Limited epidemiological data exist about amyotrophic lateral sclerosis (ALS) in the United States (US). The Agency for Toxic Substances and Disease Registry maintains the National ALS Registry and funded state and metropolitan surveillance projects to obtain reliable, timely information about ALS in defined geographic areas. METHODS Neurologists submitted case reports for ALS patients under their care between January 1, 2009 and December 31, 2011 who were New Jersey residents. A medical record verification form and electromyogram (EMG) report were requested for a sample of case reports. Incidence rates were standardized to the 2000 US Standard Population. RESULTS The average crude annual incidence rate was 1.87 per 100,000 person-years, the average age-adjusted annual incidence rate was 1.67 per 100,000 person-years, and the point prevalence rate on December 31, 2011 was 4.40 per 100,000 persons. Average annual incidence rates and point prevalence rates were statistically higher for men compared with women; Whites compared with Blacks/African Americans and Asians; and non-Hispanics compared with Hispanics. CONCLUSIONS The project findings contribute new, population-based, state-specific information to epidemiological data regarding ALS. The findings are generally consistent with previously published surveillance studies conducted in the US and abroad.
Collapse
Affiliation(s)
- Heather Jordan
- Environmental and Occupational Health Surveillance Program, New Jersey Department of Health, Trenton, N.J., USA
| | | | | | | | | |
Collapse
|
35
|
Paganoni S, Cudkowicz M, Berry JD. Outcome measures in amyotrophic lateral sclerosis clinical trials. CLINICAL INVESTIGATION 2014; 4:605-618. [PMID: 28203356 PMCID: PMC5305182 DOI: 10.4155/cli.14.52] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease with an average survival of 3-5 years. While therapies for ALS remain limited, basic and translational ALS research has been host to numerous influential discoveries in recent years. These discoveries have led to a large pipeline of potential therapies that await testing in clinical trials. Until recently, ALS clinical trials have relied on a limited cadre of 'traditional' outcome measures, including survival and measures of function. These measures have proven useful, although imperfect, in Phase III ALS trials. However, their utility in early-phase ALS trials is limited. For these early trials, outcome measures focused on target engagement or biological pathway analysis might improve trial outcomes and better support the drug development process.
Collapse
Affiliation(s)
- Sabrina Paganoni
- Harvard Medical School, Department of Neurology, Neurological Clinical Research Institute (NCRI), Massachusetts Genera Hospital, MA, USA
- Harvard Medical School, Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA
| | - Merit Cudkowicz
- Harvard Medical School, Department of Neurology, Neurological Clinical Research Institute (NCRI), Massachusetts Genera Hospital, MA, USA
| | - James D Berry
- Harvard Medical School, Department of Neurology, Neurological Clinical Research Institute (NCRI), Massachusetts Genera Hospital, MA, USA
| |
Collapse
|