1
|
Soncu Buyukiscan E, Yildirim E, Demirtas-Tatlidede A, Bilgic B, Gurvit H. An Investigation of Affective Personality Traits in Alzheimer's Disease: SEEKING as a Possible Predictor for Early-Stage Alzheimer's Dementia. Exp Aging Res 2024; 50:678-691. [PMID: 37695698 DOI: 10.1080/0361073x.2023.2256629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 09/01/2023] [Indexed: 09/13/2023]
Abstract
OBJECTIVE The aim of the current study was to investigate affective personality traits in Alzheimer's disease, a neurodegenerative condition mainly characterized by episodic memory impairment. METHOD The sample included 69 participants from 3 diagnostic categories. Twenty-five participants were diagnosed with subjective cognitive impairment (SCI), 26 participants were diagnosed with mild cognitive impairment of the amnestic type (aMCI), and the remaining 18 participants were diagnosed with early-stage Alzheimer's dementia (ADD). Diagnostic labels were given as a result of detailed neurological, neuropsychological, and neuroradiological assessment. Affective personality traits were assessed via Affective Neuroscience Personality Scales (ANPS). RESULTS The only significant intergroup difference was obtained for the SEEKING subscale of ANPS. Here, ADD group scored significantly lower compared to the SCI group. The results of logistic regression analysis also indicated that SEEKING score successfully predicted early-stage ADD diagnosis. CONCLUSION The results suggest that a specific personality constellation characterized by reduced investment in the outside world might be associated with Alzheimer's disease, either as a risk factor or a byproduct of the neurodegenerative process initiated by AD pathology.
Collapse
Affiliation(s)
| | - Elif Yildirim
- Department of Psychology, Isik University, Istanbul, Turkey
| | | | - Basar Bilgic
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hakan Gurvit
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
2
|
Lee S, Zide BS, Palm ST, Drew WJ, Sperling RA, Jacobs HIL, Siddiqi SH, Donovan NJ. Specific Association of Worry With Amyloid-β But Not Tau in Cognitively Unimpaired Older Adults. Am J Geriatr Psychiatry 2024; 32:1203-1214. [PMID: 38763835 DOI: 10.1016/j.jagp.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/21/2024]
Abstract
OBJECTIVE Anxiety disorders and subsyndromal anxiety symptoms are highly prevalent in late life. Recent studies support that anxiety may be a neuropsychiatric symptom during preclinical Alzheimer's disease (AD) and that higher anxiety is associated with more rapid cognitive decline and progression to cognitive impairment. However, the associations of specific anxiety symptoms with AD pathologies and with co-occurring subjective and objective cognitive changes have not yet been established. METHODS Baseline data from the A4 and Longitudinal Evaluation of Amyloid Risk and Neurodegeneration studies were analyzed. Older adult participants (n = 4,486) underwent assessments of anxiety (State-Trait Anxiety Inventory-6 item version [STAI]), and cerebral amyloid-beta (Aβ; 18F-florbetapir) PET and a subset underwent tau (18F-flortaucipir) PET. Linear regressions estimated associations of Aβ in a cortical composite and tau in the amygdala, entorhinal, and inferior temporal regions with STAI-Total and individual STAI item scores. Models adjusted for age, sex, education, marital status, depression, Apolipoprotein ε4 genotype, and subjective and objective cognition (Cognitive Function Index-participant; Preclinical Alzheimer Cognitive Composite). RESULTS Greater Aβ deposition was significantly associated with higher STAI-Worry, adjusting for all covariates, but not with other STAI items or STAI-Total scores. In mediation analyses, the association of Aβ with STAI-Worry was partially mediated by subjective cognition with a stronger direct effect. No associations were found for regional tau deposition with STAI-Total or STAI-Worry score. CONCLUSION Greater worry was associated with Aβ but not tau deposition, independent of subjective and objective cognition in cognitively unimpaired (CU) older adults. These findings implicate worry as an early, specific behavioral marker and a possible therapeutic target in preclinical AD.
Collapse
Affiliation(s)
- Soyoung Lee
- Division of Geriatric Psychiatry, Department of Psychiatry, Brigham and Women's Hospital (SL, BSZ, NJD), Harvard Medical School, Boston, MA; Center for Brain Circuit Therapeutics, Brigham and Women's Hospital (SL, STP, WJD, SHS), Boston, MA.
| | - Benjamin S Zide
- Division of Geriatric Psychiatry, Department of Psychiatry, Brigham and Women's Hospital (SL, BSZ, NJD), Harvard Medical School, Boston, MA
| | - Stephan T Palm
- Center for Brain Circuit Therapeutics, Brigham and Women's Hospital (SL, STP, WJD, SHS), Boston, MA; Department of Neurology, Brigham and Women's Hospital (STP, WJD, RAS, NJD), Harvard Medical School, Boston, MA
| | - William J Drew
- Center for Brain Circuit Therapeutics, Brigham and Women's Hospital (SL, STP, WJD, SHS), Boston, MA; Department of Neurology, Brigham and Women's Hospital (STP, WJD, RAS, NJD), Harvard Medical School, Boston, MA
| | - Reisa A Sperling
- Department of Neurology, Brigham and Women's Hospital (STP, WJD, RAS, NJD), Harvard Medical School, Boston, MA; Department of Neurology, Massachusetts General Hospital (RAS, NJD), Harvard Medical School, Boston, MA
| | - Heidi I L Jacobs
- Department of Radiology, Massachusetts General Hospital (HILJ), Harvard Medical School, Boston, MA; School for Mental Health and Neuroscience, Alzheimer Centre Limburg (HILJ), Maastricht University, Maastricht, The Netherlands
| | - Shan H Siddiqi
- Center for Brain Circuit Therapeutics, Brigham and Women's Hospital (SL, STP, WJD, SHS), Boston, MA; Department of Psychiatry, Brigham and Women's Hospital (SHS), Harvard Medical School, Boston, MA
| | - Nancy J Donovan
- Division of Geriatric Psychiatry, Department of Psychiatry, Brigham and Women's Hospital (SL, BSZ, NJD), Harvard Medical School, Boston, MA; Department of Neurology, Brigham and Women's Hospital (STP, WJD, RAS, NJD), Harvard Medical School, Boston, MA; Department of Psychiatry, Massachusetts General Hospital (NJD), Harvard Medical School, Boston, MA
| |
Collapse
|
3
|
Jørgensen K, Nielsen TR, Nielsen A, Oxbøll AB, Gerner SD, Waldorff FB, Waldemar G. Diagnostic accuracy of the Brief Assessment of Impaired Cognition case-finding instrument in a general practice setting and comparison with other widely used brief cognitive tests-a cross-validation study. Eur J Neurol 2024; 31:e16418. [PMID: 39045891 DOI: 10.1111/ene.16418] [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: 04/08/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND AND PURPOSE The aim of this study was to examine the discriminative validity of the Brief Assessment of Impaired Cognition (BASIC) case-finding instrument in a general practice (GP) setting and compare it with other widely used brief cognitive instruments. METHODS Patients aged ≥70 years were prospectively recruited from 14 Danish GP clinics. Participants were classified as having either normal cognition (n = 154) or cognitive impairment (n = 101) based on neuropsychological test performance, reported instrumental activities of daily living, and concern regarding memory decline. Comparisons involved the Mini-Mental State Examination (MMSE), the Montreal Cognitive Assessment (MoCA), the Rowland Universal Dementia Assessment Scale (RUDAS), the Mini-Cog, the 6-item Clock Drawing Test (CDT-6) and the BASIC Questionnaire (BASIC-Q). RESULTS BASIC demonstrated good overall classification accuracy with an area under the receiver operating characteristic curve (AUC) of 0.88 (95% confidence interval [CI] 0.84-0.92), a sensitivity of 0.72 (95% CI 0.62-0.80) and a specificity of 0.86 (95% CI 0.79-0.91). Pairwise comparisons of the AUCs of BASIC, MMSE, MoCA and RUDAS produced non-significant results, but BASIC had significantly higher classification accuracy than Mini-Cog, BASIC-Q and CDT-6. Depending on the pretest probability of cognitive impairment, the positive predictive validity of BASIC varied from 0.83 to 0.36, and the negative predictive validity from 0.97 to 0.76. CONCLUSIONS BASIC demonstrated good discriminative validity in a GP setting. The classification accuracy of BASIC is equivalent to more complex, time-consuming instruments, such as the MMSE, MoCA and RUDAS, and higher than very brief instruments, such as the CDT-6, Mini-Cog and BASIC-Q.
Collapse
Affiliation(s)
- Kasper Jørgensen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - T Rune Nielsen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Ann Nielsen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Anne-Britt Oxbøll
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Sofie D Gerner
- Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Frans B Waldorff
- Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Joyce JL, Chapman S, Waltrip L, Caes D, Gottesman R, Rizer S, Haque H, Golfer L, Mayeux RP, D'Alton ME, Marder K, Rosser M, Cosentino S. Confronting Alzheimer's Disease Risk in Women: A Feasibility Study of Memory Screening as Part of the Annual Gynecological Well-Woman Visit. J Womens Health (Larchmt) 2024; 33:1211-1218. [PMID: 38968392 DOI: 10.1089/jwh.2023.0843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024] Open
Abstract
Objective: Routine health care visits offer the opportunity to screen older adults for symptoms of Alzheimer's disease (AD). Many women see their gynecologist as their primary health care provider. Given this unique relationship, the Women's Preventive Services Initiative and the American College of Obstetrics and Gynecology advocate for integrated care of women at all ages. It is well-established that women are at increased risk for AD, and memory screening of older women should be paramount in this effort. Research is needed to determine the feasibility and value of memory screening among older women at the well-woman visit. Materials and Methods: Women aged 60 and above completed a 5-item subjective memory screener at their well-woman visit at the Columbia University Integrated Women's Health Program. Women who endorsed any item were considered to have a positive screen and were given the option to pursue clinical evaluation. Rates of positive screens, item endorsement, and referral preferences were examined. Results: Of the 530 women approached, 521 agreed to complete the screener. Of those, 17.5% (n = 91) were classified as positive. The most frequently endorsed item was difficulty with memory or thinking compared with others the same age. Among women with positive screens, 57.5% were interested in pursuing clinical referrals to a memory specialist. Conclusion: Results support the feasibility and potential value of including subjective memory screening as part of a comprehensive well-woman program. Early identification of memory loss will enable investigation into the cause of memory symptoms and longitudinal monitoring of cognitive change.
Collapse
Affiliation(s)
- Jillian L Joyce
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
| | - Silvia Chapman
- Gertrude H. Sergievsky Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Leah Waltrip
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
| | - Dorota Caes
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York, USA
| | - Reena Gottesman
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Sandra Rizer
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
| | - Hoosna Haque
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York, USA
| | - Lauren Golfer
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York, USA
| | - Richard P Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
- Gertrude H. Sergievsky Center, Columbia University Irving Medical Center, New York, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Mary E D'Alton
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York, USA
| | - Karen Marder
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
- Gertrude H. Sergievsky Center, Columbia University Irving Medical Center, New York, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Mary Rosser
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York, USA
| | - Stephanie Cosentino
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
- Gertrude H. Sergievsky Center, Columbia University Irving Medical Center, New York, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
5
|
Mann FD, Mueller AK, Zeig-Owens R, Choi J, Prezant DJ, Carr MM, Fels AM, Hennington CM, Armstrong MP, Barber A, Fontana AE, Kroll CH, Chow K, Melendez OA, Smith AJ, Luft BJ, Hall CB, Clouston SAP. Prevalence of Mild and Severe Cognitive Impairment in World Trade Center Exposed Fire Department of the City of New York (FDNY) and General Emergency Responders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.04.24311457. [PMID: 39148853 PMCID: PMC11326356 DOI: 10.1101/2024.08.04.24311457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background The emergency personnel who responded to the World Trade Center (WTC) attacks endured severe occupational exposures, yet the prevalence of cognitive impairment remains unknown among WTC-exposed-FDNY-responders. The present study screened for mild and severe cognitive impairment in WTC-exposed FDNY responders using objective tests, compared prevalence rates to a cohort of non-FDNY WTC-exposed responders, and descriptively to meta-analytic estimates of MCI from global, community, and clinical populations. Methods A sample of WTC-exposed-FDNY responders (n = 343) was recruited to complete an extensive battery of cognitive, psychological, and physical tests. The prevalences of domain-specific impairments were estimated based on the results of norm-referenced tests, and the Montreal Cognitive Assessment (MoCA), Jak/Bondi criteria, Petersen criteria, and the National Institute on Aging and Alzheimer's Association (NIA-AA) criteria were used to diagnose MCI. NIA-AA criteria were also used to diagnose severe cognitive impairment. Generalized linear models were used to compare prevalence estimates of cognitive impairment to a large sample of WTC-exposed-non-FDNY responders from the General Responder Cohort (GRC; n = 7102) who completed the MoCA during a similar time frame. Result Among FDNY responders under 65 years, the unadjusted prevalence of MCI varied from 52.57% to 71.37% depending on the operational definition of MCI, apart from using a conservative cut-off applied to MoCA total scores (18 < MoCA < 23), which yielded a markedly lower crude prevalence (24.31%) compared to alternative criteria. The prevalence of MCI was higher among WTC-exposed-FDNY-responders, compared to WTC-exposed-non-FDNY-GRC-responders (adjusted RR = 1.53, 95% C.I. = [1.24, 1.88], p < .001) and meta-analytic estimates from different global, community, and clinical populations. Following NIA-AA diagnostic guidelines, 4.96% of WTC-exposed-FDNY-responders met the criteria for severe impairments (95% CI = [2.91% to 7.82%]), a prevalence that remained largely unchanged after excluding responders over the age of 65 years. Discussion There is a high prevalence of mild and severe cognitive impairment among WTC-responders highlighting the putative role of occupational/environmental and disaster-related exposures in the etiology of accelerated cognitive decline.
Collapse
Affiliation(s)
- Frank D Mann
- Department of Family, Population, and Preventative Medicine, Program in Public Health, Renaissance School of Medicine at Stony Brook University
- Department of Medicine, Renaissance School of Medicine at Stony Brook University
| | - Alexandra K Mueller
- Bureau of Health Services, Fire Department of the City of New York, Brooklyn, NY
- Department of Medicine, Montefiore Medical Center, Bronx, NY
| | - Rachel Zeig-Owens
- Bureau of Health Services, Fire Department of the City of New York, Brooklyn, NY
- Department of Medicine, Montefiore Medical Center, Bronx, NY
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Jaeun Choi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - David J Prezant
- Bureau of Health Services, Fire Department of the City of New York, Brooklyn, NY
- Department of Medicine, Montefiore Medical Center, Bronx, NY
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Melissa M Carr
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
| | - Alicia M Fels
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
| | - Christina M Hennington
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
| | - Megan P Armstrong
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
| | - Alissa Barber
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
| | - Ashley E Fontana
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
| | - Cassandra H Kroll
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
| | - Kevin Chow
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
| | - Onix A Melendez
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
| | - Abigail J Smith
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
| | - Benjamin J Luft
- World Trade Center Health Program, Renaissance School of Medicine at Stony Brook University
- Department of Medicine, Renaissance School of Medicine at Stony Brook University
| | - Charles B Hall
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY
| | - Sean A P Clouston
- Department of Family, Population, and Preventative Medicine, Program in Public Health, Renaissance School of Medicine at Stony Brook University
| |
Collapse
|
6
|
Nakagawa S, Kowa H, Takagi Y, Kakei Y, Kagimura T, Sanada S, Nagai Y. Efficacy of a non-pharmaceutical multimodal intervention program in a group setting for patients with mild cognitive impairment: A single-arm interventional study with pre-post and external control analyses. Contemp Clin Trials Commun 2024; 40:101326. [PMID: 39021673 PMCID: PMC11252792 DOI: 10.1016/j.conctc.2024.101326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/31/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Aim This study aimed to evaluate the efficacy of a non-pharmaceutical multimodal intervention program consisting of physical exercise, cognitive stimulation, and health education in a group setting to slow the progression of mild cognitive impairment (MCI). Methods A single-arm interventional study was conducted on 27 patients with MCI. To evaluate the efficacy of the intervention program, a pre-post analysis was performed using EuroQol-5 Dimension (EQ-5D), Mini-Mental State Examination (MMSE), Cognitive Function Instrument (CFI), 5 Cog test, depression, and physical performance before and after the 8-month intervention. Additionally, propensity score and the semi-Bayes analyses were performed to compare the intervention program with standard medical care, using the external control patients' data for MMSE scores. Results Twenty-four patients completed the intervention program. During the study period, although EQ-5D and MMSE scores remained unchanged (mean change 0.02 [95 % confidence interval (CI): -0.004, 0.04], 0.5 [-0.2, 1.3]), CFI and the subcategories of 5Cog (attention and reasoning) improved (mean change -1.23 [-2.24, -0.21], 4.3 [0.9, 7.7], 3.0 [0.4, 5.6]). In the additional analysis comparing changes in MMSE scores, patients who underwent the intervention program had less decline than the external control patients (mean change -1.7 [-2.1, -1.3]) with an observed mean difference of 2.25 [1.46, 3.03], and propensity score-adjusted difference of 2.26 [1.46, 3.05]. The semi-Bayesian approach also suggested that the intervention slowed the progression of MCI. Conclusion A non-pharmaceutical multimodal intervention program could contribute to slowing cognitive decline in patients with MCI.
Collapse
Affiliation(s)
- Satoshi Nakagawa
- Division of Translational Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, Japan
| | - Hisatomo Kowa
- Division of Cognitive and Psychiatric Rehabilitation, Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Hyogo, Japan
| | - Yumi Takagi
- Department of Clinical Biostatistics, Kyoto University Graduate School of Medicine, Kyoto, Kyoto, Japan
| | - Yasumasa Kakei
- Clinical and Translational Research Centre, Kobe University Hospital, Kobe, Hyogo, Japan
| | - Tatsuo Kagimura
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, Japan
| | - Shoji Sanada
- Division of Translational Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Clinical and Translational Research Centre, Kobe University Hospital, Kobe, Hyogo, Japan
| | - Yoji Nagai
- Division of Translational Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Department of Clinical Research Facilitation Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Kyoto, Japan
| |
Collapse
|
7
|
Boza-Calvo C, Faustin A, Zhang Y, Briggs AQ, Bernard MA, Bubu OM, Rao JA, Gurin L, Tall SO, Osorio RS, Marsh K, Shao Y, Masurkar AV. Two-Year Longitudinal Outcomes of Subjective Cognitive Decline in Hispanics Compared to Non-hispanic Whites. J Geriatr Psychiatry Neurol 2024:8919887241263097. [PMID: 39043156 DOI: 10.1177/08919887241263097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
BACKGROUND Subjective cognitive decline (SCD), considered a preclinical dementia stage, is less understood in Hispanics, a high-risk group for dementia. We investigated SCD to mild cognitive impairment (MCI) progression risk, as well as baseline and longitudinal features of depressive symptoms, SCD complaints, and objective cognitive performance among Hispanics compared to non-Hispanic Whites (NHW). METHODS Hispanic (n = 23) and NHW (n = 165) SCD participants were evaluated at baseline and 2-year follow-up. Evaluations assessed function, depressive symptoms, SCD, and objective cognitive performance. RESULTS Hispanics were at increased risk of progression to MCI (OR: 6.10, 95% CI 1.09-34.20, P = .040). Hispanic participants endorsed more depressive symptoms at baseline (P = .048) that worsened more longitudinally (OR: 3.16, 95% CI 1.18-8.51, P = .023). Hispanic participants had increased SCD complaints on the Brief Cognitive Rating Scale (BCRS) (β = .40 SE: .17, P = .023), and in specific BCRS domains: concentration (β = .13, SE: .07, P = .047), past memory (β = .13, SE: .06, P = .039) and functional abilities (β = .10, SE: .05, P = .037). In objective cognitive performance, Hispanic ethnicity associated with decline in MMSE (β = -.27, SE: .13, P = .039), MoCA (β = -.80 SE: .34, P = .032), Trails A (β = 2.75, SE: .89, P = .002), Trails B (β = 9.18, SE: 2.71, P = .001) and Guild Paragraph Recall Delayed (β = -.80 SE: .28, P = .005). Conclusions: Hispanic ethnicity associated with a significantly increased risk of 2-year progression of SCD to MCI compared to NHW. This increased risk associated with increased depressive symptoms, distinctive SCD features, and elevated amnestic and non-amnestic objective cognitive decline. This supports further research to refine the assessment of preclinical dementia in this high-risk group.
Collapse
Affiliation(s)
- Carolina Boza-Calvo
- Centro de Investigación en Hematología y Trastornos Afines (CIHATA), Universidad de Costa Rica, San José, Costa Rica
- Escuela de Medicina, Universidad de Costa Rica, San José, Costa Rica
- NYU Alzheimer's Disease Research Center, NY, USA
| | - Arline Faustin
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Yian Zhang
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Anthony Q Briggs
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Mark A Bernard
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Omonigho M Bubu
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
- Center for Sleep and Brain Health, New York, NY, USA
| | - Julia A Rao
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Lindsey Gurin
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Sakina Ouedraogo Tall
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Ricardo S Osorio
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
- Center for Sleep and Brain Health, New York, NY, USA
| | - Karyn Marsh
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Yongzhao Shao
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Arjun V Masurkar
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
8
|
Yang CH, Lee J, Wilcox S, Rudisill AC, Friedman DB, Hakun JG, Neils-Strunjas J, Wei J, Miller MC, Byers MD. Implementation and evaluation of a community-based mindful walking randomized controlled trial to sustain cognitive health in older African Americans at risk for dementia. BMC Geriatr 2024; 24:579. [PMID: 38965464 PMCID: PMC11225163 DOI: 10.1186/s12877-024-05090-2] [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: 10/24/2023] [Accepted: 05/17/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND With an increasing proportion of older adults and the associated risk of Alzheimer's Disease and Related Dementias (ADRD) around the globe, there is an urgent need to engage in ADRD risk reduction efforts. African American (AA) older adults in the U.S. are disproportionally impacted by ADRD compared to other races and ethnicities. Mindful walking integrates two potentially protective factors of ADRD by elevating mindfulness and physical activity (i.e., walking), resulting in a synergistic behavioral strategy that is feasible and safe for older adults. However, the efficacy of applying this intervention for cognitive health outcomes has not been evaluated using experimental designs. METHODS This paper documents the goal and protocol of a community-based, mindful walking randomized controlled trial to examine the short- and longer-term efficacy on cognitive and other health-related outcomes in ADRD at-risk AA older adults. The study outcomes include various brain health determinants, including cognitive function, quality of life, psychological well-being, physical activity, mindfulness, sleep, and overall health status. In addition, the estimated costs of program implementation are also collected throughout the study period. This study will recruit 114 older adults (ages 60+ years) with elevated ADRD risk from the Midlands region of South Carolina. Older adults are randomly assigned to participate in 24 sessions of outdoor mindful walking over three months or a delayed mindful walking group (n=57 in each group). Participants in both groups follow identical measurement protocols at baseline, after 12 weeks, after 18 weeks, and after 24 weeks from baseline. The outcome measures are administered in the lab and in everyday settings. Costs per participant are calculated using micro-costing methods. The eliciting participant costs for mindful walking engagement with expected results are reported using the payer and the societal perspectives. DISCUSSION This study will generate evidence regarding the efficacy of mindful walking on sustaining cognitive health in vulnerable older adults. The results can inform future large-scale effectiveness trials to support our study findings. If successful, this mindful walking program can be scaled up as a low-cost and viable lifestyle strategy to promote healthy cognitive aging in diverse older adult populations, including those at greatest risk. TRIAL REGISTRATION ClinicalTrials.gov number NCT06085196 (retrospectively registered on 10/08/2023).
Collapse
Affiliation(s)
- Chih-Hsiang Yang
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA.
- Office for the Study of Aging, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA.
| | - Jongwon Lee
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Sara Wilcox
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Prevention Research Center, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - A Caroline Rudisill
- Department of Health Promotion, Education, and Behavior, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Daniela B Friedman
- Department of Health Promotion, Education, and Behavior, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Office for the Study of Aging, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Jonathan G Hakun
- Department of Neurology, The Pennsylvania State University, College of Medicine, Hershey, PA, 17033, USA
- Department of Psychology, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Public Health Sciences, The Pennsylvania State University, College of Medicine, Hershey, PA, 17033, USA
| | - Jean Neils-Strunjas
- Department of Communication Sciences and Disorders, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Office for the Study of Aging, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Jingkai Wei
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Office for the Study of Aging, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Margaret C Miller
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Office for the Study of Aging, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Megan D Byers
- Department of Health Promotion, Education, and Behavior, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Office for the Study of Aging, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
9
|
De Vito AN, Emrani S, Correia S, Coutinho MT, Lee A. Compensatory strategy use in diverse older adults with subjective cognitive complaints. Aging Ment Health 2024:1-8. [PMID: 38952264 DOI: 10.1080/13607863.2024.2367060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/06/2024] [Indexed: 07/03/2024]
Abstract
OBJECTIVES The current study aimed to evaluate the relationship between subjective cognitive complaints (SCC) and compensatory strategy (CS) use in a diverse sample of non-Latinx White (NLW), Black, and Latinx American older adults. METHOD 807 older adults (Mage = 65.38, 62.7% female) were recruited through Amazon's Mechanical Turk (MTurk) and Qualtrics Panel to complete questionnaires on SCC and CS use. Kruskall-Wallis tests were used to evaluate differences in SCC across groups given non-normal distributions. Analysis of variance (ANOVA) was used to evaluate group differences in CS use. The PROCESS macro for SPSS was used to examine whether demographic factors moderated the relationship between SCC and CS use. RESULTS NLWs reported higher levels of SCC and greater overall use of CS in comparison to Latinx and Black individuals. Several demographic and psychosocial factors including age, ethno-racial group, education, and anxiety level were found to be associated with CS use. Education was found to moderate the association between SCC and CS use. CONCLUSION Inconsistent with prior studies, our study found that NLWs reported the highest levels of SCC. CS were used across all racial/ethnic groups, but the frequency of CS use may be impacted by education level. While all education groups increased their CS in response to higher levels of SCC, this increase was more substantial for those with lower levels of education. Future work should consider individuals' cultural and educational background when examining SCC and/or developing CS-based intervention for the aging population.
Collapse
Affiliation(s)
- Alyssa N De Vito
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Butler Hospital Memory and Aging Program, Providence, RI, USA
| | - Sheina Emrani
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Butler Hospital Memory and Aging Program, Providence, RI, USA
| | - Stephen Correia
- Institute of Gerontology, University of Georgia, Athens, GA, USA
| | - Maria Teresa Coutinho
- Department of Counseling Psychology and Applied Human Development, Boston University, Boston, MA, USA
| | - Athene Lee
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Butler Hospital Memory and Aging Program, Providence, RI, USA
| |
Collapse
|
10
|
Xu L, Ren C, Jing C, Wang G, Wei H, Kong M, Ba M. Predicting amyloid-PET and clinical conversion in apolipoprotein E ε3/ε3 non-demented individuals with multidimensional factors. Eur J Neurosci 2024; 60:3742-3758. [PMID: 38698692 DOI: 10.1111/ejn.16376] [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: 01/08/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024]
Abstract
The apolipoprotein E (APOE) ε4 is a well-established risk factor of amyloid-β (Aβ) in Alzheimer's disease (AD). However, because of the high prevalence of APOE ε3, there may be a large number of people with APOE ε3/ε3 who are non-demented and have Aβ pathology. There are limited studies on assessing Aβ status and clinical conversion in the APOE ε3/ε3 non-demented population. Two hundred and ninety-three non-demented individuals with APOE ε3/ε3 from ADNI database were divided into Aβ-positron emission tomography (Aβ-PET) positivity (+) and Aβ-PET negativity (-) groups using cut-off value of >1.11. Stepwise regression searched for a single or multidimensional clinical variables for predicting Aβ-PET (+), and the receiver operating characteristic curve (ROC) assessed the accuracy of the predictive models. The Cox regression model explored the risk factors associated with clinical conversion to mild cognitive impairment (MCI) or AD. The results showed that the combination of sex, education, ventricle and white matter hyperintensity (WMH) volume can accurately predict Aβ-PET status in cognitively normal (CN), and the combination of everyday cognition study partner total (EcogSPTotal) score, age, plasma p-tau 181 and WMH can accurately predict Aβ-PET status in MCI individuals. EcogSPTotal score were independent predictors of clinical conversion to MCI or AD. The findings may provide a non-invasive and effective tool to improve the efficiency of screening Aβ-PET (+), accelerate and reduce costs of AD trial recruitment in future secondary prevention trials or help to select patients at high risk of disease progression in clinical trials.
Collapse
Affiliation(s)
- Lijuan Xu
- Department of Neurology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong, China
| | - Chao Ren
- Department of Neurology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong, China
| | - Chenxi Jing
- Department of Neurology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong, China
| | - Gang Wang
- School of Ulsan Ship and Ocean College, Ludong University, Yantai, China
| | - Hongchun Wei
- Department of Neurology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong, China
| | - Min Kong
- Department of Neurology, Yantaishan Hospital, Yantai City, Shandong, China
| | - Maowen Ba
- Department of Neurology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong, China
- Yantai Regional Sub Center of National Center for Clinical Medical Research of Neurological Diseases, Shandong, China
| |
Collapse
|
11
|
Jadick MF, Robinson T, Farrell ME, Klinger H, Buckley RF, Marshall GA, Vannini P, Rentz DM, Johnson KA, Sperling RA, Amariglio RE. Associations Between Self and Study Partner Report of Cognitive Decline With Regional Tau in a Multicohort Study. Neurology 2024; 102:e209447. [PMID: 38810211 PMCID: PMC11226320 DOI: 10.1212/wnl.0000000000209447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/04/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Self-reported cognitive decline is an early behavioral manifestation of Alzheimer disease (AD) at the preclinical stage, often believed to precede concerns reported by a study partner. Previous work shows cross-sectional associations with β-amyloid (Aβ) status and self-reported and study partner-reported cognitive decline, but less is known about their associations with tau deposition, particularly among those with preclinical AD. METHODS This cross-sectional study included participants from the Anti-Amyloid Treatment in Asymptomatic AD/Longitudinal Evaluation of Amyloid Risk and Neurodegeneration studies (N = 444) and the Harvard Aging Brain Study and affiliated studies (N = 231), which resulted in a cognitively unimpaired (CU) sample of individuals with both nonelevated (Aβ-) and elevated Aβ (Aβ+). All participants and study partners completed the Cognitive Function Index (CFI). Two regional tau composites were derived by averaging flortaucipir PET uptake in the medial temporal lobe (MTL) and neocortex (NEO). Global Aβ PET was measured in Centiloids (CLs) with Aβ+ >26 CL. We conducted multiple linear regression analyses to test associations between tau PET and CFI, covarying for amyloid, age, sex, education, and cohort. We also controlled for objective cognitive performance, measured using the Preclinical Alzheimer Cognitive Composite (PACC). RESULTS Across 675 CU participants (age = 72.3 ± 6.6 years, female = 59%, Aβ+ = 60%), greater tau was associated with greater self-CFI (MTL: β = 0.28 [0.12, 0.44], p < 0.001, and NEO: β = 0.26 [0.09, 0.42], p = 0.002) and study partner CFI (MTL: β = 0.28 [0.14, 0.41], p < 0.001, and NEO: β = 0.31 [0.17, 0.44], p < 0.001). Significant associations between both CFI measures and MTL/NEO tau PET were driven by Aβ+. Continuous Aβ showed an independent effect on CFI in addition to MTL and NEO tau for both self-CFI and study partner CFI. Self-CFI (β = 0.01 [0.001, 0.02], p = 0.03), study partner CFI (β = 0.01 [0.003, 0.02], p = 0.01), and the PACC (β = -0.02 [-0.03, -0.01], p < 0.001) were independently associated with MTL tau, but for NEO tau, PACC (β = -0.02 [-0.03, -0.01], p < 0.001) and study partner report (β = 0.01 [0.004, 0.02], p = 0.002) were associated, but not self-CFI (β = 0.01 [-0.001, 0.02], p = 0.10). DISCUSSION Both self-report and study partner report showed associations with tau in addition to Aβ. Additionally, self-report and study partner report were associated with tau above and beyond performance on a neuropsychological composite. Stratification analyses by Aβ status indicate that associations between self-reported and study partner-reported cognitive concerns with regional tau are driven by those at the preclinical stage of AD, suggesting that both are useful to collect on the early AD continuum.
Collapse
Affiliation(s)
- Michalina F Jadick
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Talia Robinson
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Michelle E Farrell
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hannah Klinger
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rachel F Buckley
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Gad A Marshall
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Patrizia Vannini
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Dorene M Rentz
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Keith A Johnson
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Reisa A Sperling
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rebecca E Amariglio
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
12
|
Liu Y, Su N, Li W, Hong B, Yan F, Wang J, Li X, Chen J, Xiao S, Yue L. Associations between Informant-Reported Cognitive Complaint and Longitudinal Cognitive Decline in Subjective Cognitive Decline A 7-Year Longitudinal Study. Arch Clin Neuropsychol 2024; 39:409-417. [PMID: 38180808 DOI: 10.1093/arclin/acad096] [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: 11/09/2023] [Revised: 11/21/2023] [Accepted: 12/07/2023] [Indexed: 01/07/2024] Open
Abstract
OBJECTIVE This study aimed to determine the predictive values of informant-reported memory decline (IMD) among subjective cognitive decline (SCD) older adults from a 7-year community-based cohort study. METHOD Ninety SCD participants were included. Demographic data and neuropsychological test scores at both baseline and 7-year follow-up were collected. Differences between SCD with IMD (+IMD) and SCD without IMD (-IMD) were compared. Logistic regression models were used to determine whether baseline IMD could predict diagnostic outcomes at 7-year follow-up. RESULTS Forty-one percent of SCD adults had IMD. At baseline, the +IMD group showed more depressive symptoms (p = 0.016) than the -IMD group. Furthermore, the Beijing-version Montreal Cognitive Assessment (MoCA), Digit Span Test-Forward, Visual Matching and Reasoning, and Wechsler Adult Intelligence Scale-RC Picture Completion (WAIS-PC) scores in the +IMD group were significantly lower than those in the -IMD group. Fifty-four percent of +IMD participants converted to mild cognitive impairment (MCI) or dementia at follow-up, and 22.6% of the -IMD participants converted to MCI. Follow-up Mini-Mental State Examination, MoCA, and Verbal Fluency Test scores of the +IMD group were significantly lower than those in the -IMD group. The +IMD group was more likely to progress to cognitive impairment at 7-year follow-up (OR = 3.361, p = 0.028). CONCLUSIONS SCD participants with +IMD may have poorer cognition and are more likely to convert to cognitive impairment over time. Our long-term follow-up study confirmed the importance of informants' perceptions of SCD, which can help clinicians identify individuals at risk of cognitive decline.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China
| | - Ning Su
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Li
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Hong
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Yan
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China
| | - Jinghua Wang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Li
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China
| | - Jianhua Chen
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shifu Xiao
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Yue
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
13
|
Schaffert J, Datoc A, Sanders GD, Didehbani N, LoBue C, Cullum CM. Repetitive head-injury exposure and later-in-life cognitive and emotional outcomes among former collegiate football players: a CLEAATS investigation. Int Rev Psychiatry 2024; 36:233-242. [PMID: 39255023 DOI: 10.1080/09540261.2024.2352572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/02/2024] [Indexed: 09/11/2024]
Abstract
This study measured the relationship between head-injury exposure and later-in-life cognitive and emotional symptoms in aging collegiate football players who participated in the College Level Aging Athlete Study. Linear regressions examined the relationship between various head-injury exposure variables (head-injury exposure estimate [HIEE], number of diagnosed concussions, and symptomatic hits to the head) and subjective cognitive function, objective cognitive function, and emotional/mood symptoms. Additional regressions evaluated the impact of emotional symptoms on subjective cognitive decline and objective cognitive function. Participants (n = 216) were 50-87 years old (M = 63.4 [8.5]), 91% White, and well-educated (bachelor's/graduate degree = 92%). HIEE did not predict scores on cognitive or emotional/mood symptom measures (p's > .169). Diagnosed concussions had a small effect on depression symptoms (p = .002, b = 0.501, R2 = .052) and subjective cognitive symptoms (p = .002, b = 0.383, R2 = .051). An emotional symptom index had a stronger relationship (p < .001, b = 0.693, R2 = .362) with subjective cognitive functioning but no significant relationship with objective cognitive function (p = .052, b = -0.211, R2 = .020). Controlling for emotional symptoms, the relationship between concussions and subjective cognitive symptoms was attenuated (p = .078, R2 = .011). Findings suggested that head-injury exposure was not significantly related to cognitive or emotional/mood outcomes in former collegiate football players and highlighted the importance of current emotional/mood symptoms on subjective cognitive function.
Collapse
Affiliation(s)
- Jeff Schaffert
- Division of Psychology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Alison Datoc
- Department of Psychiatry, Children's Health Andrews Institute, Orthopaedics and Sports Medicine, Division of Psychology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Gavin D Sanders
- Department of Psychiatry, Division of Psychology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nyaz Didehbani
- Department of Psychiatry, Physical Medicine and Rehabilitation, Division of Psychology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christian LoBue
- Department of Psychiatry, Neurological Surgery, Division of Psychology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - C Munro Cullum
- Department of Psychiatry, Neurology, and Neurological Surgery, Division of Psychology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
14
|
Mei Y, Christensen GM, Li Z, Waller LA, Ebelt S, Marcus M, Lah JJ, Wingo AP, Wingo TS, Hüls A. Joint effects of air pollution and neighborhood socioeconomic status on cognitive decline - Mediation by depression, high cholesterol levels, and high blood pressure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 923:171535. [PMID: 38453069 DOI: 10.1016/j.scitotenv.2024.171535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/21/2023] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Air pollution and neighborhood socioeconomic status (N-SES) are associated with adverse cardiovascular health and neuropsychiatric functioning in older adults. This study examines the degree to which the joint effects of air pollution and N-SES on the cognitive decline are mediated by high cholesterol levels, high blood pressure (HBP), and depression. In the Emory Healthy Aging Study, 14,390 participants aged 50+ years from Metro Atlanta, GA, were assessed for subjective cognitive decline using the cognitive function instrument (CFI). Information on the prior diagnosis of high cholesterol, HBP, and depression was collected through the Health History Questionnaire. Participants' census tracts were assigned 3-year average concentrations of 12 air pollutants and 16 N-SES characteristics. We used the unsupervised clustering algorithm Self-Organizing Maps (SOM) to create 6 exposure clusters based on the joint distribution of air pollution and N-SES in each census tract. Linear regression analysis was used to estimate the effects of the SOM cluster indicator on CFI, adjusting for age, race/ethnicity, education, and neighborhood residential stability. The proportion of the association mediated by high cholesterol levels, HBP, and depression was calculated by comparing the total and direct effects of SOM clusters on CFI. Depression mediated up to 87 % of the association between SOM clusters and CFI. For example, participants living in the high N-SES and high air pollution cluster had CFI scores 0.05 (95 %-CI:0.01,0.09) points higher on average compared to those from the high N-SES and low air pollution cluster; after adjusting for depression, this association was attenuated to 0.01 (95 %-CI:-0.04,0.05). HBP mediated up to 8 % of the association between SOM clusters and CFI and high cholesterol up to 5 %. Air pollution and N-SES associated cognitive decline was partially mediated by depression. Only a small portion (<10 %) of the association was mediated by HBP and high cholesterol.
Collapse
Affiliation(s)
- Yiyang Mei
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Grace M Christensen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Zhenjiang Li
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Lance A Waller
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Stefanie Ebelt
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Michele Marcus
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - James J Lah
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Aliza P Wingo
- Division of Mental Health, Atlanta VA Medical Center, Decatur, GA, USA; Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA
| | - Thomas S Wingo
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA; Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Anke Hüls
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
15
|
Lee Y, Nicholas ML, Connor LT. Mental Health Mediators for Subjective, Not Objective, Cognition, and Community Participation Poststroke. OTJR-OCCUPATION PARTICIPATION AND HEALTH 2024:15394492241238949. [PMID: 38494742 DOI: 10.1177/15394492241238949] [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: 03/19/2024]
Abstract
Previous studies have stated that both objective and subjective cognitive abilities and mental health symptoms are associated with community participation poststroke. However, there is a need to understand the direct and indirect associations among these variables in persons with stroke. The objective of this study was to investigate whether mental health symptoms mediate the associations of subjective and objective cognitive abilities with community participation poststroke. We built regression-based mediation models with 74 participants with mild to moderate stroke. Independent variables were objective and subjective cognitive abilities. The dependent variable was community participation. Mediators were mental health symptoms including depression, apathy, and anxiety. The results indicated that depression (b = .093), apathy (b = .134), and anxiety (b = .116) fully mediated the association between subjective cognitive ability (p < .05), but not objective cognitive ability (p > .05), and community participation poststroke. Our findings suggest that poor subjective cognitive ability combined with mental health symptoms should be addressed together to promote community participation poststroke.
Collapse
Affiliation(s)
- Yejin Lee
- Washington University School of Medicine, St. Louis, MO, USA
| | | | | |
Collapse
|
16
|
Truong QC, Choo C, Numbers K, Merkin AG, Sachdev PS, Feigin VL, Brodaty H, Kochan NA, Medvedev ON. Enhancing precision of the 16-item Informant Questionnaire on Cognitive Decline in the Elderly (IQCODE-16) using Rasch methodology. Int Psychogeriatr 2024; 36:166-176. [PMID: 34794521 DOI: 10.1017/s1041610221002568] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE This study aimed to investigate psychometric properties and enhance precision of the 16-item Informant Questionnaire on Cognitive Decline in the Elderly (IQCODE-16) up to interval-level scale using Rasch methodology. DESIGN Partial Credit Rasch model was applied to the IQCODE-16 scores using longitudinal data spanning 10 years of biennial follow-up. SETTING Community-dwelling older adults aged 70-90 years and their informants, living in Sydney, Australia, participated in the longitudinal Sydney Memory and Ageing Study (MAS). PARTICIPANTS The sample included 400 participants of the MAS aged 70 years and older, 109 out of those were diagnosed with dementia 10 years after the baseline assessment. MEASUREMENTS The IQCODE-16. RESULTS Initial analysis indicated excellent reliability of the IQCODE-16, Person Separation Index (PSI) = 0.92, but there were four misfitting items and local dependency issues. Combining locally dependent items into four super-items resulted in the best Rasch model fit with no misfitting or locally dependent items, strict unidimensionality, strong reliability, and invariance across person factors such as participants' diagnosis and relationship to their informants, as well as informants' age and sex. This permitted the generation of conversion algorithms to transform ordinal scores into interval data to enhance precision of measurement. CONCLUSIONS The IQCODE-16 demonstrated strong reliability and satisfied expectations of the unidimensional Rasch model after minor modifications. Ordinal-to-interval transformation tables published here can be used to increase accuracy of the IQCODE-16 without altering its current format. These findings could contribute to enhancement of precision in assessing clinical conditions such as cognitive decline in older people.
Collapse
Affiliation(s)
- Quoc Cuong Truong
- School of Psychology, University of Waikato, Hamilton, New Zealand
- Faculty of Psychology, Vietnam National University Ho Chi Minh City, University of Social Sciences and Humanities, Ho Chi Minh City, Vietnam
| | - Carol Choo
- College of Healthcare Sciences, Division of Tropical Health and Medicine, James Cook University, Queensland, Australia
| | - Katya Numbers
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, New South Wales, Australia
| | - Alexander G Merkin
- National Institute for Stroke and Applied Neurosciences, Auckland University of Technology, Auckland, New Zealand
- Centre for Precise Psychiatry and Neurosciences, Kaufbeuren, Germany
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, New South Wales, Australia
| | - Valery L Feigin
- National Institute for Stroke and Applied Neurosciences, Auckland University of Technology, Auckland, New Zealand
| | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, New South Wales, Australia
| | - Nicole A Kochan
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, New South Wales, Australia
| | - Oleg N Medvedev
- School of Psychology, University of Waikato, Hamilton, New Zealand
| |
Collapse
|
17
|
Huang Y, Chen H, Gao M, Lv X, Pang T, Rong S, Xu X, Yuan C. Self- and interviewer-reported cognitive problems in relation to cognitive decline and dementia: results from two prospective studies. BMC Med 2024; 22:23. [PMID: 38229039 PMCID: PMC10792911 DOI: 10.1186/s12916-023-03147-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/01/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Little is known regarding the association of interviewer-reported cognitive problems (ICP) with age-related cognitive decline. We aimed to investigate the independent associations of ICP and the combined associations of ICP and self-reported cognitive problems (SCP) with subsequent cognitive decline and dementia in two prospective cohort studies. METHODS We included 10,976 Chinese (age = 57.7 ± 8.7) and 40,499 European (age = 64.6 ± 9.4) adults without dementia from the China Health and Retirement Longitudinal Study (CHARLS) and the Survey of Health, Ageing, and Retirement in Europe (SHARE). Self-rated memory (5-point scale) and interviewer-rated frequencies of asking for clarification (6-point scale) were used to define SCP and ICP (dichotomized). Outcomes included objective cognitive test scores (z-score transformation) and incident dementia. Generalized estimating equation models were performed to evaluate mean differences in objective cognitive decline. Logistic and Cox regression models were used to estimate the relative risk of dementia. Results from two cohorts were pooled using the random-effects models. RESULTS ICP was associated with faster cognitive decline in CHARLS (βCHARLS = -0.025 [-0.044, -0.006] z-score/year). ICP and SCP were also independently associated with higher risk of dementia in two cohorts (pooled relative risk for SCP = 1.73 [1.30, 2.29]; pooled relative risk for ICP = 1.40 [1.10, 1.79]). In the joint analysis, participants with coexistence of SCP and ICP had the fastest cognitive decline (βCHARLS = -0.051 [-0.080, -0.021]; βSHARE = -0.024 [-0.043, -0.004]; pooled β = -0.035 [-0.061, -0.009] z-score/year) and highest risk of dementia (ORCHARLS = 1.77 [1.42, 2.20]; HRSHARE = 2.94 [2.42, 3.59]; pooled relative risk = 2.29 [1.38, 3.77]). CONCLUSIONS The study suggested that interviewer-reported cognitive problems may be early indicators of cognitive decline and dementia in middle-aged and older adults. A combination of self- and interviewer-reported cognitive problems showed the strongest associations with cognitive decline and dementia.
Collapse
Affiliation(s)
- Yuhui Huang
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hui Chen
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mengyan Gao
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaozhen Lv
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders, NHC Key Laboratory of Mental Health (Peking University), Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
| | - Ting Pang
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuang Rong
- Department of Nutrition and Food Hygiene, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Xin Xu
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Memory, Ageing and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Changzheng Yuan
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
18
|
Oxbøll AB, Jørgensen K, Nielsen TR, Christiansen SD, Nielsen A, Waldorff FB, Waldemar G. Diagnostic accuracy of BASIC-Q for detection of cognitive impairment in a primary care setting - a cross-validation study. BMC Geriatr 2024; 24:53. [PMID: 38212693 PMCID: PMC10785536 DOI: 10.1186/s12877-024-04675-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
OBJECTIVES This study aims to evaluate the diagnostic accuracy and reliability of a new, brief questionnaire, 'Brief Assessment of Impaired Cognition- Questionnaire' (BASIC-Q) for detection of cognitive impairment, primarily developed for use in primary care. BASIC-Q has three components: Self-report, Informant report, and Orientation. Self-report and Orientation are completed by the individual and Informant report is answered by a close relative. METHODS We included 275 participants ≥ 70 years, without a prior diagnosis of dementia, and with a close relative who agreed to participate as an informant. Participants were included prospectively in 14 general practices in urban and rural Denmark using a convenience sampling method. The Repeatable Battery for the Assessment of Neuropsychological Status (RBANS), the informant-completed Functional Activities Questionnaire (FAQ) and reported memory concern were used as a reference standard for the classification of the participants' cognitive function. RESULTS BASIC-Q demonstrated a fair to good diagnostic accuracy to differentiate between people with cognitive impairment and normal cognition with an area under the ROC curve (AUC) of 0.84 (95% CI 0.79-0.89) and a sensitivity and specificity of 0.80 (95% CI 0.72-0.87) and 0.71 (95% CI 0.63-0.78). A prorated BASIC-Q score derived from BASIC-Q without Informant report had significantly lower classification accuracy than the full BASIC-Q. The test-retest reliability of BASIC-Q was good with an intraclass correlation coefficient of 0.84. CONCLUSION BASIC-Q is a brief, easy-to-use questionnaire for identification of cognitive impairment in older adults. It demonstrated fair to good classification accuracy in a general practice setting and can be a useful case-finding tool when suspecting dementia in primary health care.
Collapse
Affiliation(s)
- Anne-Britt Oxbøll
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital- Rigshospitalet, Copenhagen, Denmark.
| | - Kasper Jørgensen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital- Rigshospitalet, Copenhagen, Denmark
| | - T Rune Nielsen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital- Rigshospitalet, Copenhagen, Denmark
| | - Sofie D Christiansen
- Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Ann Nielsen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital- Rigshospitalet, Copenhagen, Denmark
| | - Frans B Waldorff
- Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital- Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Amariglio RE, Grill JD, Rentz DM, Marshall GA, Donohue MC, Liu A, Aisen PS, Sperling RA. Longitudinal Trajectories of the Cognitive Function Index in the A4 Study. J Prev Alzheimers Dis 2024; 11:838-845. [PMID: 39044492 PMCID: PMC11266220 DOI: 10.14283/jpad.2024.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 06/18/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND The Anti-Amyloid in Asymptomatic Alzheimer's Disease (A4) Study failed to show a treatment benefit with solanezumab, but the longitudinal consequences of elevated amyloid were observed in study participants with objective decline on the Preclinical Alzheimer Cognitive Composite (PACC) and subjective decline on the combined Cognitive Function Index (participant + study partner CFI), during the trial period. OBJECTIVES We sought to expand on previous findings by comparing longitudinal patterns of participant and study partner CFI separately and their associations with the PACC stratified by baseline amyloid tertile over the course of the A4 Study. DESIGN Cognitively unimpaired older adult participants and their study partners were independently administered the CFI at screen prior to amyloid PET disclosure and then at 3 subsequent visits (week 48, week 168, week 240) of the study. PACC collected at visits concurrent with CFI administration were also examined longitudinally. SETTING The A4 Study was conducted at 67 sites in Australia, Canada, Japan, and the United States. PARTICIPANTS 1,147 participants with elevated amyloid based on florbetapir PET were enrolled in the A4 Study and included in these analyses. 583 were on placebo and 564 were treated with solanezumab. MEASUREMENTS The PACC was used to assess objective cognitive performance and the CFI was used to assess change in everyday cognitive functioning by the participant and their study partner independently. Amyloid level was characterized by Centiloid tertiles (<46.1 CL, 46.1 to 77.2 CL, >77.2 CL). Participants were aware of their elevated amyloid status, but not their CL tertile, or specific level of amyloid. Longitudinal correlations between participant and study partner CFI and PACC were examined at all visits where assessments were available. The impact of baseline amyloid tertile on CFI and PACC associations was also examined. RESULTS Both participant and study partner CFI increased over the duration of the study indicating worsening cognitive functioning. Results did not differ by treatment group. The association between higher CFI and worse PACC for both for participant and study partner became progressively stronger over the course of the study. PACC had a significantly higher correlation with study partner CFI than with participant CFI by week 168. The stronger correlations between study partner CFI and PACC were driven by those in the highest amyloid tertile. CONCLUSION Both participant and study partner report captured subtle changes in everyday cognitive functioning for participants with biomarker confirmed and disclosed preclinical AD. Moreover, study partner report was most highly aligned with cognitive decline, particularly among those with the highest amyloid load.
Collapse
Affiliation(s)
- R E Amariglio
- Rebecca E. Amariglio, Department of Neurology, Mass General Brigham, 60 Fenwood Road, Boston, MA 02115, USA, , Telephone: 617-732-9014, FAX: 617-738-9122
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Sperling RA, Donohue MC, Rissman RA, Johnson KA, Rentz DM, Grill JD, Heidebrink JL, Jenkins C, Jimenez-Maggiora G, Langford O, Liu A, Raman R, Yaari R, Holdridge KC, Sims JR, Aisen PS. Amyloid and Tau Prediction of Cognitive and Functional Decline in Unimpaired Older Individuals: Longitudinal Data from the A4 and LEARN Studies. J Prev Alzheimers Dis 2024; 11:802-813. [PMID: 39044488 PMCID: PMC11266444 DOI: 10.14283/jpad.2024.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/13/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Converging evidence suggests that markers of Alzheimer's disease (AD) pathology in cognitively unimpaired older individuals are associated with high risk of cognitive decline and progression to functional impairment. The Anti-Amyloid Treatment in Asymptomatic Alzheimer's disease (A4) and Longitudinal Evaluation of Amyloid and Neurodegeneration Risk (LEARN) Studies enrolled a large cohort of cognitively normal older individuals across a range of baseline amyloid PET levels. Recent advances in AD blood-based biomarkers further enable the comparison of baseline markers in the prediction of longitudinal clinical outcomes. OBJECTIVES We sought to evaluate whether biomarker indicators of higher levels of AD pathology at baseline predicted greater cognitive and functional decline, and to compare the relative predictive power of amyloid PET imaging, tau PET imaging, and a plasma P-tau217 assay. DESIGN All participants underwent baseline amyloid PET scan, plasma P-tau217; longitudinal cognitive testing with the Primary Alzheimer Cognitive Composite (PACC) every 6 months; and annual functional assessments with the clinical dementia rating (CDR), cognitive functional index (CFI), and activities of daily living (ADL) scales. Baseline tau PET scans were obtained in a subset of participants. Participants with elevated amyloid (Aβ+) on screening PET who met inclusion/exclusion criteria were randomized to receive placebo or solanezumab in a double-blind phase of the A4 Study over 240+ weeks. Participants who did not have elevated amyloid (Aβ-) but were otherwise eligible for the A4 Study were referred to the companion observational LEARN Study with the same outcome assessments over 240+ weeks. SETTING The A4 and LEARN Studies were conducted at 67 clinical trial sites in the United States, Canada, Japan and Australia. PARTICIPANTS Older participants (ages 65-85) who were cognitively unimpaired at baseline (CDR-GS=0, MMSE 25-30 with educational adjustment, and Logical Memory scores within the normal range LMIIa 6-18) were eligible to continue in screening. Aβ+ participants were randomized to either placebo (n=583) or solanezumab (n=564) in the A4 Study. A subset of Aβ+ underwent tau PET imaging in A4 (n=350). Aβ- were enrolled into the LEARN Study (n=553). MEASUREMENTS Baseline 18-F Florbetapir amyloid PET, 18-F Flortaucipir tau PET in a subset and plasma P-tau217 with an electrochemiluminescence (ECL) immunoassay were evaluated as predictors of cognitive (PACC), and functional (CDR, CFI and ADL) change. Models were evaluated to explore the impact of baseline tertiles of amyloid PET and tertiles of plasma P-tau217 on cognitive and functional outcomes in the A4 Study compared to LEARN. Multivariable models were used to evaluate the unique and common variance explained in longitudinal outcomes based on baseline predictors, including effects for age, gender, education, race/ethnic group, APOEε4 carrier status, baseline PACC performance and treatment assignment in A4 participants (solanezumab vs placebo). RESULTS Higher baseline amyloid PET CL and P-tau217 levels were associated with faster rates of PACC decline, and increased likelihood of progression to functional impairment (CDR 0.5 or higher on two consecutive measurements), both across LEARN Aβ- and A4 Aβ+ (solanezumab and placebo arms). In analyses considering all baseline predictor variables, P-tau217 was the strongest predictor of PACC decline. Among participants in the highest tertiles of amyloid PET or P-tau217, >50% progressed to CDR 0.5 or greater. In the tau PET substudy, neocortical tau was the strongest predictor of PACC decline, but plasma P-tau217 contributed additional independent predictive variance in commonality variance models. CONCLUSIONS In a large cohort of cognitively unimpaired individuals enrolled in a Phase 3 clinical trial and companion observational study, these findings confirm that higher baseline levels of amyloid and tau markers are associated with increased rates of cognitive decline and progression to functional impairment. Interestingly, plasma P-tau217 was the best predictor of decline in the overall sample, superior to baseline amyloid PET. Neocortical tau was the strongest predictor of cognitive decline in the subgroup with tau PET, suggesting that tau deposition is most closely linked to clinical decline. These findings indicate that biomarkers of AD pathology are useful to predict decline in an older asymptomatic population and may prove valuable in the selection of individuals for disease-modifying treatments.
Collapse
Affiliation(s)
- R A Sperling
- Reisa A. Sperling, MD, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, 617-732-8472
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Raman R, Hussen K, Donohue MC, Ernstrom K, Holdridge KC, Langford O, Molina-Henry DP, Pierce AL, Sims JR, Smith A, Yaari R, Aisen PS, Sperling R, Grill JD. Pre-Randomization Predictors of Study Discontinuation in a Preclinical Alzheimer's Disease Randomized Controlled Trial. J Prev Alzheimers Dis 2024; 11:874-880. [PMID: 39044496 PMCID: PMC11266258 DOI: 10.14283/jpad.2024.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/13/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Participant discontinuation from study treatment in a clinical trial can leave a trial underpowered, produce bias in statistical analysis, and limit interpretability of study results. Retaining participants in clinical trials for the full study duration is therefore as important as participant recruitment. OBJECTIVE This analysis aims to identify associations of pre-randomization characteristics of participants with premature discontinuation during the blinded phase of the Anti-Amyloid treatment in Asymptomatic AD (A4) Study. DESIGN All A4 trial randomized participants were classified as having prematurely discontinued study during the blinded period of the study for any reason (dropouts) or completed the blinded phase of the study on treatment (completers). SETTING The trial was conducted across 67 study sites in the United States, Canada, Japan and Australia through the global COVID-19 pandemic. PARTICIPANTS The sample consisted of all 1169 A4 trial randomized participants. MEASUREMENTS Pre-randomization demographic, clinical, amyloid PET and genetic predictors of study discontinuation were evaluated using a univariate generalized linear mixed model (GLMM), with discontinuation status as the binary outcome, each predictor as a fixed effect, and site as a random effect to account for differences among study sites in the trial. Characteristics significant at p<0.10 were then included in a multivariable GLMM. RESULTS Among randomized participants, 339 (29%) discontinued the study during the blinded period (median follow-up time in trial: 759 days). From the multivariable analysis, the two main predictors of study discontinuation were screening State-Trait Anxiety Inventory (STAI) scores (OR = 1.07 [95%CI = 1.02; 1.12]; p=0.002) and age (OR = 1.06 [95%CI = 1.03; 1.09]; p<0.001). Participants with a family history of dementia (OR = 0.75 [95%CI = 0.55; 1.01]; p=0.063) and APOE ε4 carriers (OR = 0.79 [95%CI = 0.6; 1.04]; p=0.094) were less likely to discontinue from the study, with the association being marginally significant. In these analyses, sex, race and ethnicity, cognitive scores and amyloid/tau PET scores were not associated with study dropout. CONCLUSIONS In the A4 trial, older participants and those with higher levels of anxiety at baseline as measured by the STAI were more likely to discontinue while those who had a family history of dementia or were APOE ε4 carriers were less likely to drop out. These findings have direct implications for future preclinical trial design and selection processes to identify those individuals at greatest risk of dropout and provide information to the study team to develop effective selection and retention strategies in AD prevention studies.
Collapse
Affiliation(s)
- R Raman
- Rema Raman, PhD, Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Cacciamani F, Bercu A, Bouteloup V, Grasset L, Planche V, Chêne G, Dufouil C. Understanding factors associated with the trajectory of subjective cognitive complaints in groups with similar objective cognitive trajectories. Alzheimers Res Ther 2023; 15:205. [PMID: 37993894 PMCID: PMC10666380 DOI: 10.1186/s13195-023-01348-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/05/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Cognitive complaints are often regarded as an early sign of Alzheimer's disease (AD) but may also occur in several other conditions and contexts. This study examines the correlates of cognitive complaint trajectories over a 5-year period in individuals who shared similar objective cognitive trajectories. METHODS We analyzed a subsample (n = 1748) of the MEMENTO cohort, consisting of individuals with subjective cognitive decline or mild cognitive impairment at baseline. Participants were stratified based on their latent MMSE trajectory over a 5-year period: "high and increasing," "subtle decline," and "steep decline." Within each of the three strata, we used a latent-class longitudinal approach to identify distinct trajectories of cognitive complaints. We then used multiple logistic regressions to examine the association between these complaint trajectories and several factors, including AD biomarkers (blood pTau/Aβ42 ratio, cortical thickness, APOE genotype), anxiety, depression, social relationships, a comorbidity-polypharmacy score, and demographic characteristics. RESULTS Among participants with high and increasing MMSE scores, greater baseline comorbidity-polypharmacy scores (odds ratio (OR) = 1.30, adjusted p = 0.03) were associated with higher odds of moderate and increasing cognitive complaints (as opposed to mild and decreasing complaints). Baseline depression and social relationships also showed significant associations with the complaint pattern but did not survive correction for multiple comparisons. Among participants with subtle decline in MMSE scores, greater baseline depression (OR = 1.76, adjusted p = 0.02) was associated with higher odds of moderate and increasing cognitive complaints (versus mild and decreasing). Similarly, baseline comorbidity-polypharmacy scores and pTau/Aβ42 ratio exhibited significant associations, but they did not survive correction. Among participants with a steep decline in MMSE scores, greater baseline comorbidity-polypharmacy scores increased the odds of moderate complaints (versus mild, OR = 1.38, unadjusted p = 0.03, adjusted p = 0.32), but this effect did not survive correction for multiple comparisons. CONCLUSIONS Despite similar objective cognitive trajectory, there is heterogeneity in the subjective perception of these cognitive changes. This perception was explained by both AD-related and, more robustly, non-AD-related factors. These findings deepen our understanding of the multifaceted nature of subjective cognitive complaints in individuals at risk for dementia and underscore the importance of considering a range of factors when interpreting cognitive complaints.
Collapse
Affiliation(s)
- Federica Cacciamani
- UMR 1219, Bordeaux Population Health Center, University of Bordeaux, Inserm, Bordeaux, F-33000, France.
- CIC 1401-EC, Inserm, University of Bordeaux, CHU de Bordeaux, F-33000, Bordeaux, France.
- ARAMISLab, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, CNRS, Inria, Inserm, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, F-75013, France.
- Qairnel SAS, Paris, France.
| | - Ariane Bercu
- UMR 1219, Bordeaux Population Health Center, University of Bordeaux, Inserm, Bordeaux, F-33000, France
| | - Vincent Bouteloup
- UMR 1219, Bordeaux Population Health Center, University of Bordeaux, Inserm, Bordeaux, F-33000, France
- CIC 1401-EC, Inserm, University of Bordeaux, CHU de Bordeaux, F-33000, Bordeaux, France
- Department of Public Health, CHU de Bordeaux, 33000, Bordeaux, France
| | - Leslie Grasset
- UMR 1219, Bordeaux Population Health Center, University of Bordeaux, Inserm, Bordeaux, F-33000, France
- CIC 1401-EC, Inserm, University of Bordeaux, CHU de Bordeaux, F-33000, Bordeaux, France
| | - Vincent Planche
- CHU de Bordeaux, Pôle de Neurosciences Cliniques, Centre Mémoire de Ressources Et de Recherche, 33000, Bordeaux, France
- University of Bordeaux, CNRS UMR 5293, Institut Des Maladies Neurodégénératives, 33000, Bordeaux, France
| | - Geneviève Chêne
- UMR 1219, Bordeaux Population Health Center, University of Bordeaux, Inserm, Bordeaux, F-33000, France
- CIC 1401-EC, Inserm, University of Bordeaux, CHU de Bordeaux, F-33000, Bordeaux, France
- Department of Public Health, CHU de Bordeaux, 33000, Bordeaux, France
| | - Carole Dufouil
- UMR 1219, Bordeaux Population Health Center, University of Bordeaux, Inserm, Bordeaux, F-33000, France
- CIC 1401-EC, Inserm, University of Bordeaux, CHU de Bordeaux, F-33000, Bordeaux, France
- Department of Public Health, CHU de Bordeaux, 33000, Bordeaux, France
| |
Collapse
|
23
|
Weiner MW, Aaronson A, Eichenbaum J, Kwang W, Ashford MT, Gummadi S, Santhakumar J, Camacho MR, Flenniken D, Fockler J, Truran-Sacrey D, Ulbricht A, Mackin RS, Nosheny RL. Brain health registry updates: An online longitudinal neuroscience platform. Alzheimers Dement 2023; 19:4935-4951. [PMID: 36965096 PMCID: PMC10518371 DOI: 10.1002/alz.13077] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/28/2023] [Accepted: 03/08/2023] [Indexed: 03/27/2023]
Abstract
INTRODUCTION Remote, internet-based methods for recruitment, screening, and longitudinally assessing older adults have the potential to facilitate Alzheimer's disease (AD) clinical trials and observational studies. METHODS The Brain Health Registry (BHR) is an online registry that includes longitudinal assessments including self- and study partner-report questionnaires and neuropsychological tests. New initiatives aim to increase inclusion and engagement of commonly underincluded communities using digital, community-engaged research strategies. New features include multilingual support and biofluid collection capabilities. RESULTS BHR includes > 100,000 participants. BHR has made over 259,000 referrals resulting in 25,997 participants enrolled in 30 aging and AD studies. In addition, 28,278 participants are coenrolled in BHR and other studies with data linkage among studies. Data have been shared with 28 investigators. Recent efforts have facilitated the enrollment and engagement of underincluded ethnocultural communities. DISCUSSION The major advantages of the BHR approach are scalability and accessibility. Challenges include compliance, retention, cohort diversity, and generalizability. HIGHLIGHTS Brain Health Registry (BHR) is an online, longitudinal platform of > 100,000 members. BHR made > 259,000 referrals, which enrolled 25,997 participants in 32 studies. New efforts increased enrollment and engagement of underincluded communities in BHR. The major advantages of the BHR approach are scalability and accessibility. BHR provides a unique adjunct for clinical neuroscience research.
Collapse
Affiliation(s)
- Michael W. Weiner
- Northern California Institute for Research and Education (NCIRE), Department of Veterans Affairs Medical Center, San Francisco, California, USA
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
- University of California, San Francisco Department of Psychiatry and Behavioral Sciences, San Francisco, California, USA
- University of California, San Francisco Department of Medicine, San Francisco, California, USA
- University of California, San Francisco Department of Neurology, San Francisco, California, USA
| | - Anna Aaronson
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Joseph Eichenbaum
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Winnie Kwang
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Miriam T. Ashford
- Northern California Institute for Research and Education (NCIRE), Department of Veterans Affairs Medical Center, San Francisco, California, USA
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Shilpa Gummadi
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Jessica Santhakumar
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Monica R. Camacho
- Northern California Institute for Research and Education (NCIRE), Department of Veterans Affairs Medical Center, San Francisco, California, USA
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Derek Flenniken
- Northern California Institute for Research and Education (NCIRE), Department of Veterans Affairs Medical Center, San Francisco, California, USA
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Juliet Fockler
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Diana Truran-Sacrey
- Northern California Institute for Research and Education (NCIRE), Department of Veterans Affairs Medical Center, San Francisco, California, USA
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Aaron Ulbricht
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - R. Scott Mackin
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Psychiatry and Behavioral Sciences, San Francisco, California, USA
| | - Rachel L. Nosheny
- VA Advanced Research Center, San Francisco, California, USA
- University of California, San Francisco Department of Psychiatry and Behavioral Sciences, San Francisco, California, USA
| |
Collapse
|
24
|
Sperling RA, Donohue MC, Raman R, Rafii MS, Johnson K, Masters CL, van Dyck CH, Iwatsubo T, Marshall GA, Yaari R, Mancini M, Holdridge KC, Case M, Sims JR, Aisen PS. Trial of Solanezumab in Preclinical Alzheimer's Disease. N Engl J Med 2023; 389:1096-1107. [PMID: 37458272 PMCID: PMC10559996 DOI: 10.1056/nejmoa2305032] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
BACKGROUND Trials of monoclonal antibodies that target various forms of amyloid at different stages of Alzheimer's disease have had mixed results. METHODS We tested solanezumab, which targets monomeric amyloid, in a phase 3 trial involving persons with preclinical Alzheimer's disease. Persons 65 to 85 years of age with a global Clinical Dementia Rating score of 0 (range, 0 to 3, with 0 indicating no cognitive impairment and 3 severe dementia), a score on the Mini-Mental State Examination of 25 or more (range, 0 to 30, with lower scores indicating poorer cognition), and elevated brain amyloid levels on 18F-florbetapir positron-emission tomography (PET) were enrolled. Participants were randomly assigned in a 1:1 ratio to receive solanezumab at a dose of up to 1600 mg intravenously every 4 weeks or placebo. The primary end point was the change in the Preclinical Alzheimer Cognitive Composite (PACC) score (calculated as the sum of four z scores, with higher scores indicating better cognitive performance) over a period of 240 weeks. RESULTS A total of 1169 persons underwent randomization: 578 were assigned to the solanezumab group and 591 to the placebo group. The mean age of the participants was 72 years, approximately 60% were women, and 75% had a family history of dementia. At 240 weeks, the mean change in PACC score was -1.43 in the solanezumab group and -1.13 in the placebo group (difference, -0.30; 95% confidence interval, -0.82 to 0.22; P = 0.26). Amyloid levels on brain PET increased by a mean of 11.6 centiloids in the solanezumab group and 19.3 centiloids in the placebo group. Amyloid-related imaging abnormalities (ARIA) with edema occurred in less than 1% of the participants in each group. ARIA with microhemorrhage or hemosiderosis occurred in 29.2% of the participants in the solanezumab group and 32.8% of those in the placebo group. CONCLUSIONS Solanezumab, which targets monomeric amyloid in persons with elevated brain amyloid levels, did not slow cognitive decline as compared with placebo over a period of 240 weeks in persons with preclinical Alzheimer's disease. (Funded by the National Institute on Aging and others; A4 ClinicalTrials.gov number, NCT02008357.).
Collapse
Affiliation(s)
- Reisa A Sperling
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Michael C Donohue
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Rema Raman
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Michael S Rafii
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Keith Johnson
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Colin L Masters
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Christopher H van Dyck
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Takeshi Iwatsubo
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Gad A Marshall
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Roy Yaari
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Michele Mancini
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Karen C Holdridge
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Michael Case
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - John R Sims
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Paul S Aisen
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| |
Collapse
|
25
|
Goldstein FC, Okafor M, Yang Z, Thomas T, Saleh S, Hajjar I. Subjective cognitive complaints in White and African American older adults: associations with demographic, mood, cognitive, and neuroimaging features. NEUROPSYCHOLOGY, DEVELOPMENT, AND COGNITION. SECTION B, AGING, NEUROPSYCHOLOGY AND COGNITION 2023; 30:957-970. [PMID: 37602758 PMCID: PMC10843657 DOI: 10.1080/13825585.2023.2249181] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 07/03/2023] [Indexed: 08/22/2023]
Abstract
Subjective cognitive complaints (SCC) in cognitively intact older adults have been investigated as a clinically important symptom that may portend the onset of a neurodegenerative disorder such as Alzheimer's disease. Few studies have concurrently incorporated demographic features, depressive symptoms, neuropsychological status, and neuroimaging correlates of SCC and evaluated whether these differ in White and African American older adults. In the current study, 131 (77 White, 54 African American) healthy participants ≥50 years old completed the Cognitive Function Instrument (CFI) to assess SCC, and they underwent objective cognitive testing, assessment of mood, and brain magnetic resonance imaging. Pearson Product Moment correlations were performed to evaluate associations of the CFI self-ratings with the above measures for the combined group and separately for White and African American participants. SCC were associated with greater depressive symptoms in both White and African American participants in adjusted models controlling for overall cognitive status, education, and hypertension. Greater white matter hyperintensities, lower cortical thickness, older age, and slower set shifting speed were associated with increased SCC in White participants. Although the correlations were not significant for African Americans, the strength of the associations were comparable to White participants. Hippocampal volume was not associated with either total SCC or items specific to memory functioning in the entire group. Longitudinal studies are needed to further evaluate the clinical significance of these associations with risk of conversion to mild cognitive impairment and dementia.
Collapse
Affiliation(s)
| | - Maureen Okafor
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| | - Zhiyi Yang
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| | - Tiffany Thomas
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| | - Sabria Saleh
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| | - Ihab Hajjar
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
- Department of Neurology, University of Texas Southwestern, Dallas Texas
| |
Collapse
|
26
|
Robinson T, Klinger H, Buckley R, Deters KD, Quiroz YT, Rentz D, Rabin JS, Sperling RA, Amariglio RE. Subjective cognitive decline across ethnoracial groups in the A4 study. Alzheimers Dement 2023; 19:4084-4093. [PMID: 37218387 PMCID: PMC10524317 DOI: 10.1002/alz.13138] [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: 01/03/2023] [Revised: 03/29/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023]
Abstract
INTRODUCTION The associations between subjective cognitive decline (SCD), cognition, and amyloid were explored across diverse participants in the A4 study. METHODS Five thousand one hundred and fifty-one non-Hispanic White, 262 non-Hispanic Black, 179 Hispanic-White, and 225 Asian participants completed the Preclinical Alzheimer Cognitive Composite (PACC), self- and study partner-reported Cognitive Function Index (CFI). A subsample underwent amyloid positron emission tomography (18 F-florbetapir) (N = 4384). We examined self-reported CFI, PACC, amyloid, and study partner-reported CFI by ethnoracial group. RESULTS The associations between PACC-CFI and amyloid-CFI were moderated by race. The relationships were weaker or non-significant in non-Hispanic Black and Hispanic White groups. Depression and anxiety scores were stronger predictors of CFI in these groups. Despite group differences in the types of study partners, self- and study partner-CFI were congruent across groups. DISCUSSION SCD may not uniformly relate to cognition or AD biomarkers in different ethnoracial groups. Nonetheless, self- and study partner-SCD were congruent despite differences in study partner type. Highlights Association between SCD and objective cognition was moderated by ethnoracial group. Association between SCD and amyloid was moderated by ethnoracial group. Depression and anxiety were stronger predictors of SCD in Black and Hispanic groups. Study-partner and self-reported SCD are congruent across groups. Study-partner report was consistent despite difference in study partner types.
Collapse
Affiliation(s)
- Talia Robinson
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston MA, 02115, USA
| | - Hannah Klinger
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Rachel Buckley
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston MA, 02115, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Melbourne School of Psychological Science and Florey Institute, University of Melbourne, Victoria, 3052, Australia
| | - Kacie D. Deters
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yakeel T. Quiroz
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Dorene Rentz
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston MA, 02115, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, 02115, USA
| | - Jennifer S. Rabin
- Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada, M4N 3M5
- Rehabilitation Sciences Institute, University of Toronto, Canada, M5G 1V7
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Reisa A. Sperling
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston MA, 02115, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, 02115, USA
| | - Rebecca E. Amariglio
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston MA, 02115, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, 02115, USA
| |
Collapse
|
27
|
Ahmadnezhad P, Burns JM, Akinwuntan AE, Ranchet M, Kondyli A, Mahnken JD, Devos H. Driving Automation for Older Adults with Preclinical Alzheimer's Disease. Gerontology 2023; 69:1307-1314. [PMID: 37557082 PMCID: PMC10843675 DOI: 10.1159/000531263] [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: 12/25/2022] [Accepted: 05/12/2023] [Indexed: 08/11/2023] Open
Abstract
INTRODUCTION Older adults with preclinical Alzheimer's disease (AD) show changes in on-road driving performance. The impact of preclinical AD on using automated vehicle (AV) technology is unknown. The aim was to evaluate safety and cognitive workload while operating AV technology in drivers with preclinical AD. INTRODUCTION This cross-sectional study included 40 participants: 19 older adults (age 74.16 ± 4.78; MOCA scores 26.42 ± 2.52) with preclinical AD, evidenced by elevated cortical beta-amyloid; and 21 controls (age 73.81 ± 5.62; MOCA scores 28.24 ± 1.67). All participants completed two scenarios in a driving simulator. Scenario 1 included conditional automation with an emergency event that required a manual take-over maneuver. Scenario 2 was identical but with a cognitive distractor task. Emergency response time was the main safety outcome measure. Cognitive workload was calculated using moment-to-moment changes in pupillary size and converted into an Index of Cognitive Activity (ICA). Mann-Whitney U and independent t tests were used to compare group differences. RESULTS Emergency response times were similar between drivers with preclinical AD and controls in scenario 1 (20.85 s ± 1.08 vs. 20.52 s ± 3.18; p = 0.83) and scenario 2 (14.83 s ± 7.37 vs. 13.45 s ± 10.43; p = 0.92). Likewise, no differences were found in ICA between drivers with preclinical AD and controls in scenario 1 (0.34 ± 0.08 vs. 0.33 ± 0.17; p = 0.74) or scenario 2 (0.30 ± 0.07 vs. 0.29 ± 0.17; p = 0.93). CONCLUSIONS Older drivers with preclinical AD may safely operate AV technology, without increased response times or cognitive workload. Future on-road studies with AV technology should confirm these preliminary results in drivers with preclinical AD.
Collapse
Affiliation(s)
- Pedram Ahmadnezhad
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, Kansas, USA,
| | - Jeffrey M Burns
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Abiodun E Akinwuntan
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, Kansas, USA
- University of Kansas Center for Community Access, Rehabilitation Research, Education, and Service (KU-CARES), University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Maud Ranchet
- Université Gustave Eiffel, IFSTTAR, University Lyon, Lyon, France
| | - Alexandra Kondyli
- Department of Civil, Environmental, Architectural Engineering at University of Kansas, Kansas City, Kansas, USA
| | - Jonathan D Mahnken
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hannes Devos
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, Kansas, USA
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Kansas City, Kansas, USA
- University of Kansas Center for Community Access, Rehabilitation Research, Education, and Service (KU-CARES), University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
28
|
Klinedinst BS, Kharate MK, Mohammadiarvejeh P, Fili M, Pollpeter A, Larsen BA, Moody S, Wang Q, Allenspach K, Mochel JP, Willette AA. Exploring the secrets of super-aging: a UK Biobank study on brain health and cognitive function. GeroScience 2023; 45:2471-2480. [PMID: 36947307 PMCID: PMC10651574 DOI: 10.1007/s11357-023-00765-x] [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: 01/02/2023] [Accepted: 02/27/2023] [Indexed: 03/23/2023] Open
Abstract
Communities across the globe are faced with a rapidly aging society, where age is the main risk factor for cognitive decline and development of Alzheimer's and related diseases. Despite extensive research, there have been no successful treatments yet. A rare group of individuals called "super-agers" have been noted to thrive with their exceptional ability to maintain a healthy brain and normal cognitive function even in old age. Studying their traits, lifestyles, and environments may provide valuable insight. This study used a data-driven approach to identify potential super-agers among 7121 UK Biobank participants and found that these individuals have the highest total brain volume, best cognitive performance, and lowest functional connectivity. The researchers suggest a novel hypothesis that these super-agers possess enhanced neural processing efficiency that increases with age and introduce a definition of the "neural efficiency index." Furthermore, several other types of aging were identified and significant structural-functional differences were observed between them, highlighting the benefit of research efforts in personalized medicine and precision nutrition.
Collapse
Affiliation(s)
- Brandon S Klinedinst
- Department of Medicine, University of Washington, Box 359, 325 9th Avenue, WA, 98104, Seattle, USA.
| | - Mihir K Kharate
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, USA
| | - Parvin Mohammadiarvejeh
- Department of Industrial and Manufacturing Systems Engineering, Iowa State University, Ames, IA, USA
| | - Mohammad Fili
- Department of Industrial and Manufacturing Systems Engineering, Iowa State University, Ames, IA, USA
| | - Amy Pollpeter
- Interdepartmental Bioinformatics and Computational Program, Iowa State University, Ames, IA, USA
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Brittany A Larsen
- Neuroscience Graduate Program, Iowa State University, Ames, IA, USA
- Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Shannin Moody
- Health Sciences Center, Louisiana State University, New Orleans, LA, USA
| | - Qian Wang
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, USA
| | - Jonathan P Mochel
- Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Auriel A Willette
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, USA
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
- Neuroscience Graduate Program, Iowa State University, Ames, IA, USA
- Department of Neurology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
29
|
Ng DQ, Chan D, Acharya MM, Grill JD, Chan A. Research Attitude and Interest among Cancer Survivors with or without Cognitive Impairment. Cancers (Basel) 2023; 15:3409. [PMID: 37444519 PMCID: PMC10340755 DOI: 10.3390/cancers15133409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND We examined the research attitudes and willingness to participate in clinical research among cancer survivors with varying degrees of cognitive function. METHODS This is a secondary analysis of data collected through the University of California Irvine Consent-to-Contact registry. Cancer survivors completed the Cognitive Function Instrument (CFI), the Research Attitudes Questionnaire (RAQ), and willingness to participate (WTP) in certain research procedures. Perceived cognitive impairment (CI) was defined as the worst 20% CFI scores. RESULTS Here, 265 CI and 909 cognitively non-impaired (CNI) participants' data were analyzed. Mean age and sex distribution were similar, with fewer non-Hispanic Whites and education years among CI participants. More CI participants self-reported past diagnoses of Alzheimer's disease, mild cognitive impairment, stroke, depression, post-traumatic stress disorder, and alcohol abuse (all p < 0.05). CI participants were significantly more interested in studies investigating approved medications (92% vs. 87%, p = 0.030), lumbar puncture (47% vs. 38%, p = 0.027), and autopsy (78% vs. 69%, p = 0.022). After removing survivors with co-existing neuropsychiatric conditions, interest in autopsy studies remained statistically higher among CI (79% vs. 69%, p = 0.022). CONCLUSIONS Participants with cancer and CI are open to research procedures and interventions that are traditionally less utilized, which may facilitate the discovery of the pathogenesis and interventions for cancer-related cognitive impairment (CRCI).
Collapse
Affiliation(s)
- Ding Quan Ng
- School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Daniella Chan
- School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Munjal M. Acharya
- School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Joshua D. Grill
- School of Medicine, University of California Irvine, Irvine, CA 92697, USA
- School of Biological Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Alexandre Chan
- School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
- School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
30
|
Soldevila-Domenech N, De Toma I, Forcano L, Diaz-Pellicer P, Cuenca-Royo A, Fagundo B, Lorenzo T, Gomis-Gonzalez M, Sánchez-Benavides G, Fauria K, Sastre C, Fernandez De Piérola Í, Molinuevo JL, Verdejo-Garcia A, de la Torre R. Intensive assessment of executive functions derived from performance in cognitive training games. iScience 2023; 26:106886. [PMID: 37260752 PMCID: PMC10227423 DOI: 10.1016/j.isci.2023.106886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/26/2023] [Accepted: 05/11/2023] [Indexed: 06/02/2023] Open
Abstract
Traditional neuropsychological tests accurately describe the current cognitive state but fall short to characterize cognitive change over multiple short time periods. We present an innovative approach to remote monitoring of executive functions on a monthly basis, which leverages the performance indicators from self-administered computerized cognitive training games (NUP-EXE). We evaluated the measurement properties of NUP-EXE in N = 56 individuals (59% women, 60-80 years) at increased risk of Alzheimer's disease (APOE-ϵ4 carriers with subjective cognitive decline) who completed a 12-month multimodal intervention for preventing cognitive decline. NUP-EXE presented good psychometric properties and greater sensitivity to change than traditional tests. Improvements in NUP-EXE correlated with improvements in functionality and were affected by participants' age and gender. This novel data collection methodology is expected to allow a more accurate characterization of an individual's response to a cognitive decline preventive intervention and to inform development of outcome measures for a new generation of intervention trials.
Collapse
Affiliation(s)
- Natalia Soldevila-Domenech
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Ilario De Toma
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Laura Forcano
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Patrícia Diaz-Pellicer
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Aida Cuenca-Royo
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Beatriz Fagundo
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Thais Lorenzo
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Maria Gomis-Gonzalez
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Gonzalo Sánchez-Benavides
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Karine Fauria
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | | | | | - José Luis Molinuevo
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Antonio Verdejo-Garcia
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| | - Rafael de la Torre
- Neurosciences Research Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | | |
Collapse
|
31
|
Brem AK, Kuruppu S, de Boer C, Muurling M, Diaz-Ponce A, Gove D, Curcic J, Pilotto A, Ng WF, Cummins N, Malzbender K, Nies VJM, Erdemli G, Graeber J, Narayan VA, Rochester L, Maetzler W, Aarsland D. Digital endpoints in clinical trials of Alzheimer's disease and other neurodegenerative diseases: challenges and opportunities. Front Neurol 2023; 14:1210974. [PMID: 37435159 PMCID: PMC10332162 DOI: 10.3389/fneur.2023.1210974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 05/26/2023] [Indexed: 07/13/2023] Open
Abstract
Alzheimer's disease (AD) and other neurodegenerative diseases such as Parkinson's disease (PD) and Huntington's disease (HD) are associated with progressive cognitive, motor, affective and consequently functional decline considerably affecting Activities of Daily Living (ADL) and quality of life. Standard assessments, such as questionnaires and interviews, cognitive testing, and mobility assessments, lack sensitivity, especially in early stages of neurodegenerative diseases and in the disease progression, and have therefore a limited utility as outcome measurements in clinical trials. Major advances in the last decade in digital technologies have opened a window of opportunity to introduce digital endpoints into clinical trials that can reform the assessment and tracking of neurodegenerative symptoms. The Innovative Health Initiative (IMI)-funded projects RADAR-AD (Remote assessment of disease and relapse-Alzheimer's disease), IDEA-FAST (Identifying digital endpoints to assess fatigue, sleep and ADL in neurodegenerative disorders and immune-mediated inflammatory diseases) and Mobilise-D (Connecting digital mobility assessment to clinical outcomes for regulatory and clinical endorsement) aim to identify digital endpoints relevant for neurodegenerative diseases that provide reliable, objective, and sensitive evaluation of disability and health-related quality of life. In this article, we will draw from the findings and experiences of the different IMI projects in discussing (1) the value of remote technologies to assess neurodegenerative diseases; (2) feasibility, acceptability and usability of digital assessments; (3) challenges related to the use of digital tools; (4) public involvement and the implementation of patient advisory boards; (5) regulatory learnings; and (6) the significance of inter-project exchange and data- and algorithm-sharing.
Collapse
Affiliation(s)
- Anna-Katharine Brem
- Department of Old Age Psychiatry, King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
- University Hospital of Old Age Psychiatry, University of Bern, Bern, Switzerland
| | - Sajini Kuruppu
- Department of Old Age Psychiatry, King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
| | - Casper de Boer
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
| | - Marijn Muurling
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
| | | | | | - Jelena Curcic
- Novartis Institutes for Biomedical Research (NIBR), Basel, Switzerland
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Wan-Fai Ng
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- NIHR Newcastle Biomedical Research Centre and Clinical Research Facility, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Nicholas Cummins
- Department of Biostats and Health Informatics, King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
| | | | | | - Gul Erdemli
- Novartis Pharmaceuticals Corporations, Cambridge, MA, United States
| | - Johanna Graeber
- Institute of General Practice, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
| | | | - Lynn Rochester
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- National Institute for Health and Care Research (NIHR) Newcastle Biomedical Research Centre (BRC), Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Walter Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein and Kiel University, Kiel, Germany
| | - Dag Aarsland
- Department of Old Age Psychiatry, King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
32
|
Rafii MS, Aisen PS. Detection and treatment of Alzheimer's disease in its preclinical stage. NATURE AGING 2023; 3:520-531. [PMID: 37202518 PMCID: PMC11110912 DOI: 10.1038/s43587-023-00410-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/29/2023] [Indexed: 05/20/2023]
Abstract
Longitudinal multimodal biomarker studies reveal that the continuum of Alzheimer's disease (AD) includes a long latent phase, referred to as preclinical AD, which precedes the onset of symptoms by decades. Treatment during the preclinical AD phase offers an optimal opportunity for slowing the progression of disease. However, trial design in this population is complex. In this Review, we discuss the recent advances in accurate plasma measurements, new recruitment approaches, sensitive cognitive instruments and self-reported outcomes that have facilitated the successful launch of multiple phase 3 trials for preclinical AD. The recent success of anti-amyloid immunotherapy trials in symptomatic AD has increased the enthusiasm for testing this strategy at the earliest feasible stage. We provide an outlook for standard screening of amyloid accumulation at the preclinical stage in clinically normal individuals, during which effective therapy to delay or prevent cognitive decline can be initiated.
Collapse
Affiliation(s)
- Michael S Rafii
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine University of Southern California, Los Angeles, CA, USA.
| | - Paul S Aisen
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
33
|
Li C, Hong Y, Yang X, Zeng X, Ocepek-Welikson K, Eimicke JP, Kong J, Sano M, Zhu C, Neugroschl J, Aloysi A, Cai D, Martin J, Loizos M, Sewell M, Akrivos J, Evans K, Sheppard F, Greenberg J, Ardolino A, Teresi JA. The use of subjective cognitive complaints for detecting mild cognitive impairment in older adults across cultural and linguistic groups: A comparison of the Cognitive Function Instrument to the Montreal Cognitive Assessment. Alzheimers Dement 2023; 19:1764-1774. [PMID: 36222321 PMCID: PMC10090224 DOI: 10.1002/alz.12804] [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: 04/14/2022] [Revised: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 11/11/2022]
Abstract
INTRODUCTION This pilot study aims to explore the psychometric properties of the Cognitive Function Instrument (CFI) as a measure of subjective cognitive complaints (SCC) and its performance in distinguishing mild cognitive impairment (MCI) from normal control (NC) compared to an objective cognitive screen (Montreal Cognitive Assessment [MoCA]). METHODS One hundred ninety-four community-dwelling non-demented older adults with racial/ethnic diversity were included. Unidimensionality and internal consistency of the CFI were examined using factor analysis, Cronbach's alpha, and McDonald's omega. Logistic regression models and receiver operating characteristic (ROC) analysis were used to examine the performance of CFI. RESULTS The CFI demonstrated adequate internal consistency; however, the fit for a unidimensional model was suboptimal. The CFI distinguished MCI from NC alone or in combination with MoCA. ROC analysis showed comparable performance of the CFI and the MoCA. DISCUSSION Our findings support the use of CFI as a brief and easy-to-use screen to detect MCI in culturally/linguistically diverse older adults. HIGHLIGHT What is the key scientific question or problem of central interest of the paper? Subjective cognitive complaints (SCCs) are considered the earliest sign of dementia in older adults. However, it is unclear if SCC are equivalent in different cultures. The Cognitive Function Instrument (CFI) is a 14-item measure of SCC. This study provides pilot data suggesting that CFI is sensitive for detecting mild cognitive impairment in a cohort of older adults with racial/ethnic diversity. Comparing performance, CFI demonstrates comparable sensitivity to the Montreal Cognitive Assessment, an objective cognitive screening test. Overall, SCC may provide a non-invasive, easy-to-use method to flag possible cognitive impairment in both research and clinical settings.
Collapse
Affiliation(s)
- Clara Li
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yue Hong
- Salem Hospital, Mass General Brigham, Salem, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Xiao Yang
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xiaoyi Zeng
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katja Ocepek-Welikson
- Columbia University Stroud Center, New York State Psychiatric Institute, New York, NY, USA
| | | | - Jian Kong
- Research Division, The Hebrew Home at Riverdale, Riverdale, NY, USA
| | - Mary Sano
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, New York, NY, USA
| | - Carolyn Zhu
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, New York, NY, USA
| | - Judith Neugroschl
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amy Aloysi
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dongming Cai
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, New York, NY, USA
| | - Jane Martin
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Loizos
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margaret Sewell
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jimmy Akrivos
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kirsten Evans
- James J. Peters VA Medical Center, New York, NY, USA
| | - Faye Sheppard
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Greenberg
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Allison Ardolino
- Alzheimer’s Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeanne A. Teresi
- Columbia University Stroud Center, New York State Psychiatric Institute, New York, NY, USA
- Research Division, The Hebrew Home at Riverdale, Riverdale, NY, USA
- Mount Sinai Pepper Older Americans Independence Center, Department of Geriatrics and Palliative Medicine, Mount Sinai Medical Center, New York, NY, USA
| |
Collapse
|
34
|
Rabin LA, Sikkes SA, Tommet D, Jones RN, Crane PK, Elbulok-Charcape MM, Dubbelman MA, Koscik R, Amariglio RE, Buckley RF, Boada M, Chételat G, Dubois B, Ellis KA, Gifford KA, Jefferson AL, Jessen F, Johnson S, Katz MJ, Lipton RB, Luck T, Margioti E, Maruff P, Molinuevo JL, Perrotin A, Petersen RC, Rami L, Reisberg B, Rentz DM, Riedel-Heller SG, Risacher SL, Rodriguez-Gomez O, Sachdev PS, Saykin AJ, Scarmeas N, Smart C, Snitz BE, Sperling RA, Taler V, van der Flier WM, van Harten AC, Wagner M, Wolfsgruber S. Linking self-perceived cognitive functioning questionnaires using item response theory: The subjective cognitive decline initiative. Neuropsychology 2023; 37:463-499. [PMID: 37276136 PMCID: PMC10564559 DOI: 10.1037/neu0000888] [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] [Indexed: 06/07/2023] Open
Abstract
OBJECTIVE Self-perceived cognitive functioning, considered highly relevant in the context of aging and dementia, is assessed in numerous ways-hindering the comparison of findings across studies and settings. Therefore, the present study aimed to link item-level self-report questionnaire data from international aging studies. METHOD We harmonized secondary data from 24 studies and 40 different questionnaires with item response theory (IRT) techniques using a graded response model with a Bayesian estimator. We compared item information curves to identify items with high measurement precision at different levels of the self-perceived cognitive functioning latent trait. Data from 53,030 neuropsychologically intact older adults were included, from 13 English language and 11 non-English (or mixed) language studies. RESULTS We successfully linked all questionnaires and demonstrated that a single-factor structure was reasonable for the latent trait. Items that made the greatest contribution to measurement precision (i.e., "top items") assessed general and specific memory problems and aspects of executive functioning, attention, language, calculation, and visuospatial skills. These top items originated from distinct questionnaires and varied in format, range, time frames, response options, and whether they captured ability and/or change. CONCLUSIONS This was the first study to calibrate self-perceived cognitive functioning data of geographically diverse older adults. The resulting item scores are on the same metric, facilitating joint or pooled analyses across international studies. Results may lead to the development of new self-perceived cognitive functioning questionnaires guided by psychometric properties, content, and other important features of items in our item bank. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
Affiliation(s)
- Laura A. Rabin
- Department of Psychology, Brooklyn College, Brooklyn, NY, USA and The Graduate Center of CUNY, NY, NY, USA
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sietske A.M. Sikkes
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Douglas Tommet
- Department of Psychiatry and Human Behavior and Neurology, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Richard N. Jones
- Department of Psychiatry and Human Behavior and Neurology, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Paul K. Crane
- Division of General Internal Medicine, University of Washington, Seattle, WA, USA
| | | | - Mark A. Dubbelman
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Rebecca Koscik
- Wisconsin Alzheimer's Institute University of Wisconsin School of Medicine and Public Health Madison WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health Madison WI, USA
| | - Rebecca E. Amariglio
- Department of Neurology, Brigham and Women’s Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachel F. Buckley
- Department of Neurology, Brigham and Women’s Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry and Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Australia
| | - Mercè Boada
- Alzheimer Research Center and Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Gaël Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", NeuroPresage Team, Cyceron, 14000 Caen, France
| | - Bruno Dubois
- Université Pierre et Marie Curie-Paris 6, AP-HP, Hôpital de la Salpêtrière, Paris, France
- Centre des Maladies Cognitives et Comportementales, Institut du Cerveau et de la Moelle épinière (ICM), UMRS975, Paris, France
| | - Kathryn A. Ellis
- Department of Psychiatry and Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Australia
| | - Katherine A. Gifford
- Vanderbilt Memory & Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Angela L. Jefferson
- Vanderbilt Memory & Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Sterling Johnson
- Geriatric Research Education and Clinical Center William S. Middleton Memorial Veterans Hospital Madison WI, USA
- University of Wisconsin School of Medicine and Public Health, Madison WI, USA
| | - Mindy J. Katz
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Richard B. Lipton
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Psychiatry and Behavioral Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tobias Luck
- Faculty of Applied Social Sciences, University of Applied Sciences Erfurt, Erfurt, Germany
| | - Eleni Margioti
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Aviv Clinics, Jumeirah Lake Towers, Dubai, United Arab Emirates
| | | | - Jose Luis Molinuevo
- Alzheimer’s Disease and Other Cognitive Disorders Unit, IDIBAPS, Hospital Clinic, Barcelona, Spain
| | - Audrey Perrotin
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", NeuroPresage Team, Cyceron, 14000 Caen, France
| | - Ronald C. Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Lorena Rami
- Alzheimer’s Disease and Other Cognitive Disorders Unit, IDIBAPS, Hospital Clinic, Barcelona, Spain
| | - Barry Reisberg
- Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
- Silberstein Aging and Dementia Research Center, New York University School of Medicine, New York, NY, USA
| | - Dorene M. Rentz
- Department of Neurology, Brigham and Women’s Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steffi G. Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health (ISAP), University of Leipzig, Leipzig, Germany
| | - Shannon L. Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN
| | - Octavio Rodriguez-Gomez
- Alzheimer Research Center and Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades, Barcelona, Spain
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Greece
- Department of Neurology, Columbia University, New York, USA
| | - Colette Smart
- Department of Psychology, University of Victoria, Victoria, BC, Canada
- Centre on Aging, University of Victoria, Victoria, BC, Canada
| | - Beth E. Snitz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Reisa A. Sperling
- Department of Neurology, Brigham and Women’s Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vanessa Taler
- School of Psychology, University of Ottawa, Ottawa, ON, Canada
- Bruyère Research Institute, Ottawa, ON, Canada
| | - Wiesje M. van der Flier
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Argonde C. van Harten
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry and Psychotherapy University of Bonn, Bonn, Germany
| | | |
Collapse
|
35
|
Chandler JM, Rentz DM, Zagar A, Kim Y, Schwartz RL, Fillit H. Disease progression and costs at the 3-year follow-up of the GERAS-US study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12430. [PMID: 37091310 PMCID: PMC10113935 DOI: 10.1002/dad2.12430] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/15/2023] [Accepted: 03/19/2023] [Indexed: 04/25/2023]
Abstract
Introduction GERAS-US prospectively characterized clinical and economic outcomes of early symptomatic Alzheimer's disease (AD). Societal cost changes were examined in amyloid-positive patients with mild cognitive impairment due to AD (MCI) and mild dementia due to AD (MILD). Methods Cognition, function, and caregiver burden were assessed using Mini-Mental State Examination (MMSE), Cognitive Function Index (CFI), and Zarit Burden Interview, respectively. Costs are presented as least square mean for the overall population and for MCI versus MILD using mixed model repeated measures. Results MMSE score and CFI worsened. Total societal costs (dollars/month) for MCI and MILD, respectively, were higher at baseline ($2430 and $4063) but steady from 6 ($1977 and $3032) to 36 months ($2007 and $3392). Direct non-medical costs rose significantly for MILD. Caregiver burden was higher for MILD versus MCI at 12, 18, and 24 months. Discussion Function and cognition declined in MILD. Non-medical costs reflect the increasing impact of AD even in its early stages. HIGHLIGHTS In the GERAS-US study, total societal costs for patients with mild cognitive impairment due to Alzheimer's disease (MCI) and mild dementia due to Alzheimer's disease (MILD) were higher at baseline but steady from 6 to 36 months.Mini-Mental State Examination (MMSE) and Cognitive Function Index (CFI) worsened; the rate of decline was significant for patients with MILD but not for those with MCI.There was a rise in direct non-medical costs at 36 months for patients with MILD.Caregiver burden was higher for MILD versus MCI at 12, 18, and 24 months.Slowing the rate of disease progression in this early symptomatic population may allow patients to maintain their ability to carry out everyday activities longer.
Collapse
Affiliation(s)
| | - Dorene M. Rentz
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyBrigham and Women's HospitalMassachusetts General HospitalBostonMassachusettsUSA
| | | | - Yongin Kim
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | | | - Howard Fillit
- Geriatric Medicine, Palliative Care and NeuroscienceThe Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Alzheimer's Drug Discovery FoundationNew YorkNew YorkUSA
| |
Collapse
|
36
|
Reynolds G, Buckley R, Papp K, Schultz SA, Rentz D, Sperling R, Amariglio R. Relation of modifiable lifestyle and mood factors to cognitive concerns among participants and their study partners in the A4 screen data. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12435. [PMID: 37304049 PMCID: PMC10248212 DOI: 10.1002/dad2.12435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/16/2023] [Accepted: 04/10/2023] [Indexed: 06/13/2023]
Abstract
Introduction Subjective cognitive decline (SCD) has been associated with elevated amyloid levels and increased risk of future cognitive decline, as well as modifiable variables, including depression, anxiety, and physical inactivity. Participants generally endorse greater and earlier concerns than their close family and friends (study partners [SPs]), which may reflect subtle changes at the earliest stages of disease among participants with underlying neurodegenerative processes. However, many individuals with subjective concerns are not at risk of Alzheimer's disease (AD) pathology, suggesting that additional factors, such as lifestyle habits, may be contributory. Methods We examined the relation between SCD, amyloid status, lifestyle habits (exercise, sleep), mood/anxiety, and demographic variables among 4481 cognitively unimpaired older adults who are being screened for a multi-site secondary prevention trial (A4 screen data; mean ±SD: age = 71.3 ±4.7, education = 16.6 ±2.8, 59% women, 96% non-Hispanic or Latino, 92% White]. Results On the Cognitive Function Index (CFI) participants endorsed higher concerns compared to SPs. Participant concerns were associated with older age, positive amyloid status, worse mood/anxiety, lower education, and lower exercise, whereas SP concerns were associated with older participant age, male gender of participant, positive amyloid status of participant, and worse participant-reported mood/anxiety. Discussion Findings suggest that modifiable/lifestyle factors (e.g., exercise, education) may be associated with participant concerns among cognitively unimpaired individuals and highlight the importance of further examining how modifiable factors impact participant- and SP-reported concerns, which may inform trial recruitment and clinical interventions.
Collapse
Affiliation(s)
- Gretchen Reynolds
- Department of NeurologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Rachel Buckley
- Department of NeurologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Melbourne School of Psychological SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| | - Kathryn Papp
- Department of NeurologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Stephanie A. Schultz
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Dorene Rentz
- Department of NeurologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Reisa Sperling
- Department of NeurologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Rebecca Amariglio
- Department of NeurologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
37
|
Braley TJ, Shieu MM, Zaheed AB, Dunietz GL. Pathways between multiple sclerosis, sleep disorders, and cognitive function: Longitudinal findings from The Nurses' Health Study. Mult Scler 2023; 29:436-446. [PMID: 36633265 PMCID: PMC9991978 DOI: 10.1177/13524585221144215] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND The potential mediating and moderating effects of sleep disorders on cognitive outcomes in multiple sclerosis (MS) have been insufficiently studied. OBJECTIVES To determine direct and indirect longitudinal associations between sleep disorders and perceived cognitive dysfunction in women with MS. METHODS The 2013 and 2017 waves of the Nurses' Health Study (n = 63,866) were utilized. All diagnoses and symptoms including MS (n = 524) were self-reported. Subjective cognitive function was measured using a composite score of four memory items and three binary outcomes that assessed difficulty following instructions, conversations/plots, and street navigation. Moderating and mediating effects of diagnosed/suspected obstructive sleep apnea (OSA), sleepiness, and insomnia between MS and cognition were estimated using the four-way decomposition method. RESULTS Prevalence of diagnosed/suspected OSA, sleepiness, and insomnia in 2013 were higher for nurses with MS (NwMS). NwMS were more likely to report cognitive difficulties in 2017. Insomnia mediated 5.4%-15.1% of the total effect between MS and following instructions, conversations/plots, and memory impairment, while sleepiness mediated 8.6%-12.3% of the total effect for these outcomes. In interaction analyses, OSA significantly accounted for 34% of the total effect between MS and following instructions. CONCLUSION Prevalent OSA, insomnia, and sleepiness could differentially moderate or mediate the effect of MS on cognition in women with MS.
Collapse
Affiliation(s)
- Tiffany J Braley
- Division of Multiple Sclerosis and Clinical Neuroimmunology, Department of Neurology, University of Michigan, Ann Arbor, MI, USA/Division of Sleep Medicine, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Monica M Shieu
- Division of Sleep Medicine, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Afsara B Zaheed
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Galit Levi Dunietz
- Division of Sleep Medicine, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
38
|
Shand C, Markiewicz PJ, Cash DM, Alexander DC, Donohue MC, Barkhof F, Oxtoby NP. Heterogeneity in Preclinical Alzheimer's Disease Trial Cohort Identified by Image-based Data-Driven Disease Progression Modelling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.07.23285572. [PMID: 36798314 PMCID: PMC9934776 DOI: 10.1101/2023.02.07.23285572] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Importance Undetected biological heterogeneity adversely impacts trials in Alzheimer's disease because rate of cognitive decline - and perhaps response to treatment - differs in subgroups. Recent results show that data-driven approaches can unravel the heterogeneity of Alzheimer's disease progression. The resulting stratification is yet to be leveraged in clinical trials. Objective Investigate whether image-based data-driven disease progression modelling could identify baseline biological heterogeneity in a clinical trial, and whether these subgroups have prognostic or predictive value. Design Screening data from the Anti-Amyloid Treatment in Asymptomatic Alzheimer Disease (A4) Study collected between April 2014 and December 2017, and longitudinal data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) observational study downloaded in February 2022 were used. Setting The A4 Study is an interventional trial involving 67 sites in the US, Canada, Australia, and Japan. ADNI is a multi-center observational study in North America. Participants Cognitively unimpaired amyloid-positive participants with a 3-Tesla T1-weighted MRI scan. Amyloid positivity was determined using florbetapir PET imaging (in A4) and CSF Aβ(1-42) (in ADNI). Main Outcomes and Measures Regional volumes estimated from MRI scans were used as input to the Subtype and Stage Inference (SuStaIn) algorithm. Outcomes included cognitive test scores and SUVr values from florbetapir and flortaucipir PET. Results We included 1,240 Aβ+ participants (and 407 Aβ- controls) from the A4 Study, and 731 A4-eligible ADNI participants. SuStaIn identified three neurodegeneration subtypes - Typical, Cortical, Subcortical - comprising 523 (42%) individuals. The remainder are designated subtype zero (insufficient atrophy). Baseline PACC scores (A4 primary outcome) were significantly worse in the Cortical subtype (median = -1.27, IQR=[-3.34,0.83]) relative to both subtype zero (median=-0.013, IQR=[-1.85,1.67], P<.0001) and the Subcortical subtype (median=0.03, IQR=[-1.78,1.61], P=.0006). In ADNI, over a four-year period (comparable to A4), greater cognitive decline in the mPACC was observed in both the Typical (-0.23/yr; 95% CI, [-0.41,-0.05]; P=.01) and Cortical (-0.24/yr; [-0.42,-0.06]; P=.009) subtypes, as well as the CDR-SB (Typical: +0.09/yr, [0.06,0.12], P<.0001; and Cortical: +0.07/yr, [0.04,0.10], P<.0001). Conclusions and Relevance In a large secondary prevention trial, our image-based model detected neurodegenerative heterogeneity predictive of cognitive heterogeneity. We argue that such a model is a valuable tool to be considered in future trial design to control for previously undetected variance.
Collapse
Affiliation(s)
- Cameron Shand
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Pawel J Markiewicz
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - David M Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Daniel C Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Michael C Donohue
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, USA
| | - Frederik Barkhof
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
| | - Neil P Oxtoby
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| |
Collapse
|
39
|
Chang HT, Chiu PY. Development of a simple screening tool for determining cognitive status in Alzheimer's disease. PLoS One 2023; 18:e0280178. [PMID: 36634049 PMCID: PMC9836308 DOI: 10.1371/journal.pone.0280178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Cognitive screening is often a first step to document cognitive status of patients suspected having Alzheimer's disease (AD). Unfortunately, screening neuropsychological tests are often insensitivity in the detection. The goal of this study was to develop a simple and sensitive screening neuropsychological test to facilitate early detection of AD. This study recruited 761 elderly individuals suspected of having AD and presenting various cognitive statuses (mean age: 77.69 ± 8.45 years; proportion of females: 65%; cognitively unimpaired, CU, n = 133; mild cognitive impairment, MCI, n = 231; dementia of Alzheimer's type, DAT, n = 397). This study developed a novel screening neuropsychological test incorporating assessments of the core memory deficits typical of early AD and an interview on memory function with an informant. The proposed History-based Artificial Intelligence-Show Chwan Assessment of Cognition (HAI-SAC) was assessed in terms of psychometric properties, test time, and discriminative ability. The results were compared with those obtained using other common screening tests, including Cognitive Abilities Screening Instrument (CASI), Montreal Cognitive Assessment (MoCA), and an extracted Mini-Mental State Examination score from CASI. HAI-SAC demonstrated acceptable internal consistency. Factor analysis revealed two factors: memory (semantic and contextual) and cognition-related information from informants. The assessment performance of HAI-SAC was strongly correlated with that of the common screening neuropsychological tests addressed in this study. HAI-SAC outperformed the other tests in differentiating CU individuals from patients with MCI (sensitivity: 0.87; specificity: 0.58; area under the curve [AUC]: 0.78) or DAT (sensitivity: 0.99; specificity: 0.89; AUC: 0.98). Performance of HAI-SAC on differentiating MCI from DAT was on par with performances of other tests (sensitivity: 0.78; specificity: 0.84; AUC: 0.87), while the test time was less than one quarter that of CASI and half that of MoCA. HAI-SAC is psychometrically sound, cost-effective, and sensitive in discriminating the cognitive status of AD.
Collapse
Affiliation(s)
- Hsin-Te Chang
- Department of Psychology, College of Science, Chung Yuan Christian University, Taoyuan, Taiwan
- Research Assistance Center, Show Chwan Memorial Hospital, Changhua City, Taiwan
| | - Pai-Yi Chiu
- Department of Neurology, Show Chwan Memorial Hospital, Changhua City, Taiwan
- Department of Applied Mathematics, College of Science, Tunghai University, Taichung, Taiwan
| |
Collapse
|
40
|
Kumagai R, Osaki T, Oki Y, Murata S, Uchida K, Encho H, Ono R, Kowa H. The Japan-Multimodal Intervention Trial for Prevention of Dementia PRIME Tamba (J-MINT PRIME Tamba): Study protocol of a randomised controlled multi-domain intervention trial. Arch Gerontol Geriatr 2023; 104:104803. [PMID: 36088747 DOI: 10.1016/j.archger.2022.104803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 02/07/2023]
Abstract
The Japan-Multimodal Intervention Trial for Prevention of Dementia PRIME Tamba (J-MINT PRIME Tamba) is a randomised controlled trial to prevent cognitive decline in community-dwelling cognitively ordinary older people at risk of dementia. Participants are aged 65-85 years living in a rural area in Japan, aware of very mild decline in cognitive function or abilities of activities of daily living, have at least one vascular risk (e.g. hypertension or diabetes), and have a Mini-Mental State Examination score of 24 or higher. Approximately 200 participants are randomly divided into two groups, with the intervention group receiving a multi-modal intervention, including lifestyle-related disease management, physical exercise, cognitive training, and nutritional counselling, over 18 months. The primary outcome is change in the composite score of seven neuropsychological tests, including the Free and Cued Selective Reminding Test, Logical Memory I and II subsets of the Wechsler Memory Scale-Revised, and Digit Symbol Substitution Test of the Wechsler Adult Intelligence Scale. In addition, changes in a wide range of other parameters such as physical function, blood test results, sleep, and frailty are also analysed as secondary outcomes. We believe that this study's results will contribute significantly to the development of dementia prevention measures in Japan. Clinical trial registration number: UMIN000041938.
Collapse
Affiliation(s)
- Ryoko Kumagai
- Division of Cognitive and Psychiatric Rehabilitation, Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan
| | - Tohmi Osaki
- Centre for Preventing Dementia, Kobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan; Department of Occupational Therapy, Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani-cho, Nishi-ku, Kobe, Hyogo 651-2180, Japan.
| | - Yutaro Oki
- Centre for Preventing Dementia, Kobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan
| | - Shunsuke Murata
- Centre for Preventing Dementia, Kobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan; Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibeshimmachi, Suita, Osaka 564-8565, Japan
| | - Kazuaki Uchida
- Division of Community Health Sciences, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan; Department of Prevention and Care Science, Research Institute, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi 474-8511, Japan
| | - Haruhi Encho
- Division of Community Health Sciences, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan
| | - Rei Ono
- Division of Community Health Sciences, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan; Department of Physical Activity Research, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, 1-23-1, Toyama, Shinjyuku, Tokyo 162-8636, Japan
| | - Hisatomo Kowa
- Division of Cognitive and Psychiatric Rehabilitation, Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, 7-10-2, Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan.
| |
Collapse
|
41
|
Numbers K, Lam BCP, Crawford JD, Kochan NA, Sachdev PS, Brodaty H. Longitudinal changes in participant and informant reports of subjective cognitive complaints are associated with dementia risk. Front Aging Neurosci 2023; 15:1044807. [PMID: 36891557 PMCID: PMC9987247 DOI: 10.3389/fnagi.2023.1044807] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/27/2023] [Indexed: 02/22/2023] Open
Abstract
Background Individuals with subjective cognitive complaints (SCCs) are at an increased risk of dementia. Questions remain about participant-reported versus informant-reported SCCs as indicators of future dementia and about longitudinal changes in participant-and informant-reported SCCs and risk of incident dementia. Method Participants were 873 older adults (M = 78.65-years; 55% female) and 849 informants from the Sydney Memory and Ageing Study. Comprehensive assessments occurred biennially, and clinical diagnoses were made by expert consensus for 10-years. SCCs were participants' and informants' responses to a single binary question concerning their/the participant's memory decline (Yes/No) over the first 6-years. Categorical latent growth curve analyses, using the logit transformation, were used to model SCC change over time. Associations of initial propensity to report SCCs at baseline, and change in propensity to report SCCs over time, with dementia risk were examined using Cox regression. Results 70% of participants reported SCCs at baseline, with a proportional increase in the odds of reporting by 11% for each additional year in the study. In contrast, 22% of informants reported SCCs at baseline, with a proportional increase by 30% in the odds of reporting per year. Participants' initial level of (p = 0.007), but not change in SCC reporting (p = 0.179), was associated with risk of dementia controlling for all covariates. Both informants' initial level of (p < 0.001), and change in (p < 0.001), SCCs significantly predicted incident dementia. When modelled together, informants' initial level of, and change in, SCCs were still independently associated with increased dementia risk (p's < 0.001). Conclusion These data suggest that informants' initial impressions, and increased reporting, of SCCs appear to be uniquely prognostic of future dementia compared to participants', even based on a single SCC question.
Collapse
Affiliation(s)
- Katya Numbers
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Ben C P Lam
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - John D Crawford
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Nicole A Kochan
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
42
|
Pérez-Blanco L, Felpete A, Patten SB, Mallo SC, Pereiro AX, Campos-Magdaleno M, Juncos-Rabadán O. Do informant-reported subjective cognitive complaints predict progression to mild cognitive impairment and dementia better than self-reported complaints in old adults? A meta-analytical study. Ageing Res Rev 2022; 82:101772. [PMID: 36374732 DOI: 10.1016/j.arr.2022.101772] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 10/05/2022] [Accepted: 10/27/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Subjective cognitive complaints (SCCs) are considered a risk factor for objective cognitive decline and conversion to dementia. The aim of this study was to determine whether self-reported or informant-reported SCCs best predict progression to mild cognitive impairment (MCI) and/or dementia. METHODS We reviewed prospective longitudinal studies of Cognitively Unimpaired (CU) older adults with self-reported and informant-reported SCCs at baseline, assessed by questions or questionnaires that considered the transition to MCI and/or dementia. A random-effects meta-analysis was performed to obtain pooled estimates and 95% CIs. RESULTS Both self-reported and informant-reported SCCs are associated with an elevated risk of transition from CU to MCI and/or dementia. The association appears stronger and more robust for informant-reported data [1.38, with a 95% CI of 1.16 -1.64, p < 0.001] than for self-reported data [1.27 (95% CI 1.06 - 1.534, p = 0.011]. CONCLUSIONS Our results suggest that corroborated information from one informant could provide important details for distinguishing between normal aging and clinical states.
Collapse
Affiliation(s)
- Lucía Pérez-Blanco
- Department of Developmental Psychology, Universidade de Santiago de Compostela, Xosé María Suárez Núñez Street, Campus Sur, Santiago de Compostela, Galicia ES 15782, Spain.
| | - Alba Felpete
- Department of Developmental Psychology, Universidade de Santiago de Compostela, Xosé María Suárez Núñez Street, Campus Sur, Santiago de Compostela, Galicia ES 15782, Spain
| | - Scott B Patten
- Department of Psychiatry, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada
| | - Sabela C Mallo
- Department of Developmental Psychology, Universidade de Santiago de Compostela, Xosé María Suárez Núñez Street, Campus Sur, Santiago de Compostela, Galicia ES 15782, Spain
| | - Arturo X Pereiro
- Department of Developmental Psychology, Universidade de Santiago de Compostela, Xosé María Suárez Núñez Street, Campus Sur, Santiago de Compostela, Galicia ES 15782, Spain
| | - María Campos-Magdaleno
- Department of Developmental Psychology, Universidade de Santiago de Compostela, Xosé María Suárez Núñez Street, Campus Sur, Santiago de Compostela, Galicia ES 15782, Spain
| | - Onésimo Juncos-Rabadán
- Department of Developmental Psychology, Universidade de Santiago de Compostela, Xosé María Suárez Núñez Street, Campus Sur, Santiago de Compostela, Galicia ES 15782, Spain
| |
Collapse
|
43
|
Maffoni M, Pierobon A, Fundarò C. MASCoD-Multidimensional Assessment of Subjective Cognitive Decline. Front Psychol 2022; 13:921062. [PMID: 36533024 PMCID: PMC9748696 DOI: 10.3389/fpsyg.2022.921062] [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: 04/15/2022] [Accepted: 05/30/2022] [Indexed: 09/19/2023] Open
Abstract
Subjective cognitive decline (SCD) is a subclinical cognitive impairment that is complained by the individual without being objectively supported at clinical, diagnostic, and neuropsychological levels. It can negatively impact on patient's frailty and quality of life, as well as on the caregiver's burden. Moreover, it can be prodromal to Mild Cognitive Impairment or dementia. Although the clinical manifestations of SCD can differ along with several cognitive domains, to date there are only screening tools to investigate subjective memory complaints. Thus, the first aim of this paper is to propose a preliminary English and Italian version of a new screening tool called MASCoD (Multidimensional Assessment of Subjective Cognitive Decline); the second aim is to propose its preliminary adoption on a pilot sample. This schedule is a brief test derived from the review of the literature and the clinical experience provided by an experts panelist. From pilot tests, it seems promising as it can help the professional to make differential diagnosis and to predict the risk of developing severe cognitive impairment over time, developing a personalized care path. This screening tool is brief, easily embeddable in usual clinical assessment, and administrable by different professionals. Furthermore, following validation, it will allow to collect manifold cognitive manifestations of SCD, addressing the shortage of previous validated instruments globally assessing cognition affected by this condition.
Collapse
Affiliation(s)
- Marina Maffoni
- Psychology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Montescano Institute, Montescano, Italy
| | - Antonia Pierobon
- Psychology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Montescano Institute, Montescano, Italy
| | - Cira Fundarò
- Neurophysiopatology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Montescano Institute, Montescano, Italy
| |
Collapse
|
44
|
Cattaneo G, Pachón-García C, Roca A, Alviarez-Schulze V, Opisso E, García-Molina A, Bartrés-Faz D, Pascual-Leone A, Tormos-Muñoz JM, Solana-Sánchez J. "Guttmann Cognitest" ®, preliminary validation of a digital solution to test cognitive performance. Front Aging Neurosci 2022; 14:987891. [PMID: 36408102 PMCID: PMC9669647 DOI: 10.3389/fnagi.2022.987891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/10/2022] [Indexed: 02/19/2024] Open
Abstract
Thanks to technological advances, the administration of cognitive assessments via digital solutions continues to increase, both in research and clinical practice. "Guttmann Cognitest"°ledR is a digital solution for cognitive assessment which includes seven computerized tasks designed to assess main cognitive functions requiring approximately 20 min to be completed. The purpose of the present study was to validate it against standard and more extensive in-person neuropsychological assessments in the context of the Barcelona Brain Health Initiative (BBHI) cohort study. We studied 274 participants of the BBHI (126 women, mean age = 56.14, age range 44-69), who underwent an extensive in-person assessment, including a classical paper-and-pencil neuropsychological assessment and a cognitive assessment via the "Guttmann Cognitest"°ledR. Principal component analysis indicated that "Guttmann Cognitest"°ledR measures four main cognitive domains and convergent validity analysis demonstrated that cognitive performance was associated with gold standard paper and pencil tests. Results also showed an expected negative correlation with age, a relation with educational level as well as a gender effect. Regression-based norming equations for the sample tested are also reported. Performing a cognitive assessment with this digital solution is feasible and potentially useful to gather information about cognitive functioning in large samples and experimental settings.
Collapse
Affiliation(s)
- Gabriele Cattaneo
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
| | - Catherine Pachón-García
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
| | - Alba Roca
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Barcelona, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Vanessa Alviarez-Schulze
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- Departamento de Ciencias del Comportamiento, Escuela de Psicología, Universidad Metropolitana, Caracas, Venezuela
| | - Eloy Opisso
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
| | - Alberto García-Molina
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
| | - David Bartrés-Faz
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Barcelona, Spain
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Alvaro Pascual-Leone
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Barcelona, Spain
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, United States
- Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Josep M. Tormos-Muñoz
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
| | - Javier Solana-Sánchez
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la UAB, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Barcelona, Spain
| |
Collapse
|
45
|
Van Dyk K, Ahn J, Zhou X, Zhai W, Ahles TA, Bethea TN, Carroll JE, Cohen HJ, Dilawari AA, Graham D, Jacobsen PB, Jim H, McDonald BC, Nakamura ZM, Patel SK, Rentscher KE, Saykin AJ, Small BJ, Mandelblatt JS, Root JC. Associating persistent self-reported cognitive decline with neurocognitive decline in older breast cancer survivors using machine learning: The Thinking and Living with Cancer study. J Geriatr Oncol 2022; 13:1132-1140. [PMID: 36030173 PMCID: PMC10016202 DOI: 10.1016/j.jgo.2022.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 07/16/2022] [Accepted: 08/10/2022] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Many cancer survivors report cognitive problems following diagnosis and treatment. However, the clinical significance of patient-reported cognitive symptoms early in survivorship can be unclear. We used a machine learning approach to determine the association of persistent self-reported cognitive symptoms two years after diagnosis and neurocognitive test performance in a prospective cohort of older breast cancer survivors. MATERIALS AND METHODS We enrolled breast cancer survivors with non-metastatic disease (n = 435) and age- and education-matched non-cancer controls (n = 441) between August 2010 and December 2017 and followed until January 2020; we excluded women with neurological disease and all women passed a cognitive screen at enrollment. Women completed the FACT-Cog Perceived Cognitive Impairment (PCI) scale and neurocognitive tests of attention, processing speed, executive function, learning, memory and visuospatial ability, and timed activities of daily living assessments at enrollment (pre-systemic treatment) and annually to 24 months, for a total of 59 individual neurocognitive measures. We defined persistent self-reported cognitive decline as clinically meaningful decline (3.7+ points) on the PCI scale from enrollment to twelve months with persistence to 24 months. Analysis used four machine learning models based on data for change scores (baseline to twelve months) on the 59 neurocognitive measures and measures of depression, anxiety, and fatigue to determine a set of variables that distinguished the 24-month persistent cognitive decline group from non-cancer controls or from survivors without decline. RESULTS The sample of survivors and controls ranged in age from were ages 60-89. Thirty-three percent of survivors had self-reported cognitive decline at twelve months and two-thirds continued to have persistent decline to 24 months (n = 60). Least Absolute Shrinkage and Selection Operator (LASSO) models distinguished survivors with persistent self-reported declines from controls (AUC = 0.736) and survivors without decline (n = 147; AUC = 0.744). The variables that separated groups were predominantly neurocognitive test performance change scores, including declines in list learning, verbal fluency, and attention measures. DISCUSSION Machine learning may be useful to further our understanding of cancer-related cognitive decline. Our results suggest that persistent self-reported cognitive problems among older women with breast cancer are associated with a constellation of mild neurocognitive changes warranting clinical attention.
Collapse
Affiliation(s)
- Kathleen Van Dyk
- Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, United States of America; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, United States of America.
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University, Washington, DC, United States of America
| | - Xingtao Zhou
- Georgetown Lombardi Comprehensive Cancer Center Georgetown University, Washington, DC, United States of America
| | - Wanting Zhai
- Georgetown Lombardi Comprehensive Cancer Center Georgetown University, Washington, DC, United States of America
| | - Tim A Ahles
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Traci N Bethea
- Georgetown Lombardi Comprehensive Cancer Center Georgetown University, Washington, DC, United States of America
| | - Judith E Carroll
- Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, United States of America; Cousins Center for Psychoneuroimmunology, University of California, Los Angeles, CA, United States of America
| | - Harvey Jay Cohen
- Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC, United States of America
| | - Asma A Dilawari
- Georgetown Lombardi Comprehensive Cancer Center Georgetown University, Washington, DC, United States of America
| | - Deena Graham
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, United States of America
| | - Paul B Jacobsen
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, United States of America
| | - Heather Jim
- Department of Health Outcomes and Behavior, Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL, United States of America
| | - Brenna C McDonald
- Center for Neuroimaging, Department of Radiology and Imaging Sciences and the Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Zev M Nakamura
- Department of Psychiatry, University of North Carolina-Chapel Hill, Chapel Hill, NC, United States of America
| | - Sunita K Patel
- City of Hope National Medical Center, Los Angeles, CA, United States of America
| | - Kelly E Rentscher
- Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, United States of America; Cousins Center for Psychoneuroimmunology, University of California, Los Angeles, CA, United States of America
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences and the Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Brent J Small
- University of South Florida, Health Outcome and Behavior Program and Biostatistics Resource Core, H. Lee Moffitt Cancer Center, Research Institute at the University of South Florida, Tampa, FL, United States of America
| | - Jeanne S Mandelblatt
- Georgetown Lombardi Comprehensive Cancer Center Georgetown University, Washington, DC, United States of America
| | - James C Root
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| |
Collapse
|
46
|
Nosheny RL, Amariglio R, Sikkes SA, Van Hulle C, Bicalho MAC, Dowling NM, Brucki SMD, Ismail Z, Kasuga K, Kuhn E, Numbers K, Aaronson A, Moretti DV, Pereiro AX, Sánchez‐Benavides G, Sellek Rodríguez AF, Urwyler P, Zawaly K. The role of dyadic cognitive report and subjective cognitive decline in early ADRD clinical research and trials: Current knowledge, gaps, and recommendations. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12357. [PMID: 36226046 PMCID: PMC9530696 DOI: 10.1002/trc2.12357] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/05/2022] [Accepted: 08/22/2022] [Indexed: 11/05/2022]
Abstract
Efficient identification of cognitive decline and Alzheimer's disease (AD) risk in early stages of the AD disease continuum is a critical unmet need. Subjective cognitive decline is increasingly recognized as an early symptomatic stage of AD. Dyadic cognitive report, including subjective cognitive complaints (SCC) from a participant and an informant/study partner who knows the participant well, represents an accurate, reliable, and efficient source of data for assessing risk. However, the separate and combined contributions of self- and study partner report, and the dynamic relationship between the two, remains unclear. The Subjective Cognitive Decline Professional Interest Area within the Alzheimer's Association International Society to Advance Alzheimer's Research and Treatment convened a working group focused on dyadic patterns of subjective report. Group members identified aspects of dyadic-report information important to the AD research field, gaps in knowledge, and recommendations. By reviewing existing data on this topic, we found evidence that dyadic measures are associated with objective measures of cognition and provide unique information in preclinical and prodromal AD about disease stage and progression and AD biomarker status. External factors including dyad (participant-study partner pair) relationship and sociocultural factors contribute to these associations. We recommend greater dyad report use in research settings to identify AD risk. Priority areas for future research include (1) elucidation of the contributions of demographic and sociocultural factors, dyad type, and dyad relationship to dyad report; (2) exploration of agreement and discordance between self- and study partner report across the AD syndromic and disease continuum; (3) identification of domains (e.g., memory, executive function, neuropsychiatric) that predict AD risk outcomes and differentiate cognitive impairment due to AD from other impairment; (4) development of best practices for study partner engagement; (5) exploration of study partner report as AD clinical trial endpoints; (6) continued development, validation, and optimization, of study partner report instruments tailored to the goals of the research and population.
Collapse
Affiliation(s)
- Rachel L. Nosheny
- University of California San FranciscoDepartment of PsychiatrySan FranciscoCaliforniaUSA
- Veteran's Administration Advanced Research CenterSan FranciscoCaliforniaUSA
| | - Rebecca Amariglio
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalDepartment of Neurology Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Sietske A.M. Sikkes
- Amsterdam University Medical CentersDepartment of NeurologyAlzheimer Center AmsterdamNorth Hollandthe Netherlands/VU UniversityDepartment of ClinicalNeuro & Development PsychologyNorth Hollandthe Netherlands
| | - Carol Van Hulle
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Maria Aparecida Camargos Bicalho
- UFMG: Federal University of Minas GeraisDepartment of Clinical MedicineJenny de Andrade Faria – Center for Geriatrics and Gerontology of UFMGBelo HorizonteBrazil
| | - N. Maritza Dowling
- George Washington UniversityDepartment of Acute & Chronic CareSchool of NursingDepartment of Epidemiology & BiostatisticsMilken Institute School of Public HealthWashingtonDistrict of ColumbiaUSA
| | | | - Zahinoor Ismail
- Hotchkiss Brain Institute and O'Brien Institute for Public HealthCumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Kensaku Kasuga
- Department of Molecular GeneticsBrain Research InstituteNiigata UniversityNiigataJapan
| | - Elizabeth Kuhn
- UNICAEN, INSERM, PhIND “Physiopathology and Imaging of Neurological Disorders,”Institut Blood and Brain @ Caen‐NormandieNormandie UniversityCaenFrance
| | - Katya Numbers
- Centre for Healthy Brain Ageing (CHeBA)Department of PsychiatryUniversity of New South WalesSydneyNew South WalesAustralia
| | - Anna Aaronson
- Veteran's Administration Advanced Research CenterSan FranciscoCaliforniaUSA
| | - Davide Vito Moretti
- IRCCS Istituto Centro San Giovanni di Dio FatebenefratelliAlzheimer Rehabilitation Operative UnitBresciaItaly
| | - Arturo X. Pereiro
- Faculty of PsychologyDepartment of Developmental PsychologyUniversity of Santiago de CompostelaGaliciaSpain
| | | | - Allis F. Sellek Rodríguez
- Costa Rican Foundation for the Care of Older Adults with Alzheimer's and Other Dementias (FundAlzheimer Costa Rica)CartagoCosta Rica
| | - Prabitha Urwyler
- ARTORG Center for Biomedical EngineeringUniversity of BernUniversity Neurorehabilitation UnitDepartment of NeurologyInselspitalBernSwitzerland
| | - Kristina Zawaly
- University of AucklandDepartment of General Practice and Primary Health CareSchool of Population HealthFaculty of Medical and Health SciencesAucklandNew Zealand
| | | |
Collapse
|
47
|
Li Z, Christensen GM, Lah JJ, Marcus M, Russell AG, Ebelt S, Waller LA, Hüls A. Neighborhood characteristics as confounders and effect modifiers for the association between air pollution exposure and subjective cognitive functioning. ENVIRONMENTAL RESEARCH 2022; 212:113221. [PMID: 35378125 PMCID: PMC9233127 DOI: 10.1016/j.envres.2022.113221] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 05/25/2023]
Abstract
BACKGROUND Air pollution has been associated with cognitive function in the elderly. Previous studies have not evaluated the simultaneous effect of neighborhood-level socioeconomic status (N-SES), which can be an essential source of bias. OBJECTIVES We explored N-SES as a confounder and effect modifier in a cross-sectional study of air pollution and subjective cognitive function. METHODS We included 12,058 participants age 50+ years from the Emory Healthy Aging Study in Metro Atlanta using the Cognitive Function Instrument (CFI) score as our outcome, with higher scores representing worse subjective cognitive function. We estimated 9-year average ambient carbon monoxide (CO), nitrogen oxides (NOx), and fine particulate matter (PM2.5) concentrations at residential addresses using a fusion of dispersion and chemical transport models. We collected census-tract level N-SES indicators and created two composite measures via principal component analysis and k-means clustering. Associations between pollutants and CFI and effect modification by N-SES were estimated via linear regression models adjusted for age, education, race and N-SES. RESULTS N-SES confounded the association between air pollution and CFI, independent of individual characteristics. We found significant effect modifications by N-SES for the association between air pollution and CFI (p-values<0.001) suggesting that effects of air pollution differ depending on N-SES. Participants living in areas with low N-SES were most vulnerable to air pollution. In the lowest N-SES urban areas, interquartile range (IQR) increases in CO, NOx, and PM2.5 were associated with 5.4% (95%-confidence interval, -0.2,11.3), 4.9% (-0.4,10.4), and 9.8% (2.2,18.0) changes in CFI, respectively. In lowest N-SES suburban areas, IQR increases in CO, NOx, and PM2.5 were associated with higher changes in CFI, namely 13.0% (0.9,26.5), 13.0% (-0.1,27.8), and 17.3% (2.5,34.2), respectively. DISCUSSION N-SES is an important confounder and effect modifier in our study. This finding could have implications for studying health effects of air pollution and identifying susceptible populations.
Collapse
Affiliation(s)
- Zhenjiang Li
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Grace M Christensen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - James J Lah
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Michele Marcus
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Armistead G Russell
- School of Civil and Environmental Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Stefanie Ebelt
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Lance A Waller
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Anke Hüls
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
48
|
Christensen GM, Li Z, Pearce J, Marcus M, Lah JJ, Waller LA, Ebelt S, Hüls A. The complex relationship of air pollution and neighborhood socioeconomic status and their association with cognitive decline. ENVIRONMENT INTERNATIONAL 2022; 167:107416. [PMID: 35868076 PMCID: PMC9382679 DOI: 10.1016/j.envint.2022.107416] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 06/13/2023]
Abstract
BACKGROUND Air pollution and neighborhood socioeconomic status (nSES) have been shown to affect cognitive decline in older adults. In previous studies, nSES acts as both a confounder and an effect modifier between air pollution and cognitive decline. OBJECTIVES This study aims to examine the individual and joint effects of air pollution and nSES on cognitive decline on adults 50 years and older in Metro Atlanta, USA. METHODS Perceived memory and cognitive decline was assessed in 11,897 participants aged 50+ years from the Emory Healthy Aging Study (EHAS) using the cognitive function instrument (CFI). Three-year average air pollution concentrations for 12 pollutants and 16 nSES characteristics were matched to participants using census tracts. Individual exposure linear regression and LASSO models explore individual exposure effects. Environmental mixture modeling methods including, self-organizing maps (SOM), Bayesian kernel machine regression (BKMR), and quantile-based G-computation explore joint effects, and effect modification between air pollutants and nSES characteristics on cognitive decline. RESULTS Participants living in areas with higher air pollution concentrations and lower nSES experienced higher CFI scores (beta: 0.121; 95 % CI: 0.076, 0.167) compared to participants living in areas with low air pollution and high nSES. Additionally, the BKMR model showed a significant overall mixture effect on cognitive decline, suggesting synergy between air pollution and nSES. These joint effects explain protective effects observed in single-pollutant linear regression models, even after adjustment for confounding by nSES (e.g., an IQR increase in CO was associated with a 0.038-point lower (95 % CI: -0.06, -0.01) CFI score). DISCUSSION Observed protective effects of single air pollutants on cognitive decline can be explained by joint effects and effect modification of air pollutants and nSES. Researchers must consider nSES as an effect modifier if not a co-exposure to better understand the complex relationships between air pollution and nSES in urban settings.
Collapse
Affiliation(s)
- Grace M Christensen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Zhenjiang Li
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - John Pearce
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Michele Marcus
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - James J Lah
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Lance A Waller
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Stefanie Ebelt
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Anke Hüls
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
49
|
Bernstein OM, Vegetabile BG, Salazar CR, Grill JD, Gillen DL. Adjustment for biased sampling using NHANES derived propensity weights. HEALTH SERVICES AND OUTCOMES RESEARCH METHODOLOGY 2022. [DOI: 10.1007/s10742-022-00283-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
50
|
Aisen PS, Jimenez-Maggiora GA, Rafii MS, Walter S, Raman R. Early-stage Alzheimer disease: getting trial-ready. Nat Rev Neurol 2022; 18:389-399. [PMID: 35379951 PMCID: PMC8978175 DOI: 10.1038/s41582-022-00645-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 12/15/2022]
Abstract
Slowing the progression of Alzheimer disease (AD) might be the greatest unmet medical need of our time. Although one AD therapeutic has received a controversial accelerated approval from the FDA, more effective and accessible therapies are urgently needed. Consensus is growing that for meaningful disease modification in AD, therapeutic intervention must be initiated at very early (preclinical or prodromal) stages of the disease. Although the methods for such early-stage clinical trials have been developed, identification and recruitment of the required asymptomatic or minimally symptomatic study participants takes many years and requires substantial funds. As an example, in the Anti-Amyloid Treatment in Asymptomatic Alzheimer’s Disease Trial (the first phase III trial to be performed in preclinical AD), 3.5 years and more than 5,900 screens were required to recruit and randomize 1,169 participants. A new clinical trials infrastructure is required to increase the efficiency of recruitment and accelerate therapeutic progress. Collaborations in North America, Europe and Asia are now addressing this need by establishing trial-ready cohorts of individuals with preclinical and prodromal AD. These collaborations are employing innovative methods to engage the target population, assess risk of brain amyloid accumulation, select participants for biomarker studies and determine eligibility for trials. In the future, these programmes could provide effective tools for pursuing the primary prevention of AD. Here, we review the lessons learned from the AD trial-ready cohorts that have been established to date, with the aim of informing ongoing and future efforts towards efficient, cost-effective trial recruitment. Consensus is growing that intervention in the very early stages of Alzheimer disease is necessary for disease modification. Here, the authors discuss the challenges of recruiting asymptomatic or mildly symptomatic participants for clinical trials, focusing on ‘trial-ready’ cohorts as a potential solution. Trial-ready cohorts are an effective strategy for the identification of participants eligible for clinical trials in early-stage Alzheimer disease (AD). Building these cohorts requires considerable planning and technological infrastructure to facilitate recruitment, remote longitudinal assessment, data management and data storage. Trial-ready cohorts exist for genetically determined populations at risk of AD, such as those with familial AD and Down syndrome; the longitudinal data from these cohorts is improving our understanding of the disease progression in early stages, informing clinical trial design and accelerating recruitment to intervention studies. So far, the challenges experienced by trial-ready cohorts for early-stage AD have included difficulties recruiting an ethnically and racially representative cohort; and for online cohorts, difficulty retaining participants. The results of ongoing work will reveal the success of strategies to improve cohort diversity and retention, and the rates of referral to clinical trials.
Collapse
|