Hostname: page-component-7bb8b95d7b-cx56b Total loading time: 0 Render date: 2024-09-26T21:58:56.112Z Has data issue: false hasContentIssue false

Comparing neuropsychological, typical, and ADNI criteria for the diagnosis of mild cognitive impairment in Vietnam-era veterans

Published online by Cambridge University Press:  24 January 2024

Monica T. Ly*
Affiliation:
Veterans Affairs San Diego Healthcare System, San Diego, CA, USA Department of Psychiatry, University of California San Diego Health, La Jolla, CA, USA
Jennifer Adler
Affiliation:
Veterans Affairs San Diego Healthcare System, San Diego, CA, USA Department of Psychiatry, University of California San Diego Health, La Jolla, CA, USA
Adan F. Ton Loy
Affiliation:
Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
Emily C. Edmonds
Affiliation:
Banner Alzheimer’s Institute, Tucson, AZ, USA Departments of Neurology and Psychology, University of Arizona, Tucson, AZ, USA
Mark W. Bondi
Affiliation:
Veterans Affairs San Diego Healthcare System, San Diego, CA, USA Department of Psychiatry, University of California San Diego Health, La Jolla, CA, USA
Lisa Delano-Wood
Affiliation:
Veterans Affairs San Diego Healthcare System, San Diego, CA, USA Department of Psychiatry, University of California San Diego Health, La Jolla, CA, USA Center for Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
*
Corresponding author: M. Ly; Email: monicaly@gmail.com
Rights & Permissions [Opens in a new window]

Abstract

Objective:

Neuropsychological criteria for mild cognitive impairment (MCI) more accurately predict progression to Alzheimer’s disease (AD) and are more strongly associated with AD biomarkers and neuroimaging profiles than ADNI criteria. However, research to date has been conducted in relatively healthy samples with few comorbidities. Given that history of traumatic brain injury (TBI) and post-traumatic stress disorder (PTSD) are risk factors for AD and common in Veterans, we compared neuropsychological, typical (Petersen/Winblad), and ADNI criteria for MCI in Vietnam-era Veterans with histories of TBI or PTSD.

Method:

267 Veterans (mean age = 69.8) from the DOD-ADNI study were evaluated for MCI using neuropsychological, typical, and ADNI criteria. Linear regressions adjusting for age and education assessed associations between MCI status and AD biomarker levels (cerebrospinal fluid [CSF] p-tau181, t-tau, and Aβ42) by diagnostic criteria. Logistic regressions adjusting for age and education assessed the effects of TBI severity and PTSD symptom severity simultaneously on MCI classification by each criteria.

Results:

Agreement between criteria was poor. Neuropsychological criteria identified more Veterans with MCI than typical or ADNI criteria, and were associated with higher CSF p-tau181 and t-tau. Typical and ADNI criteria were not associated with CSF biomarkers. PTSD symptom severity predicted MCI diagnosis by neuropsychological and ADNI criteria. History of moderate/severe TBI predicted MCI by typical and ADNI criteria.

Conclusions:

MCI diagnosis using sensitive neuropsychological criteria is more strongly associated with AD biomarkers than conventional diagnostic methods. MCI diagnostics in Veterans would benefit from incorporation of comprehensive neuropsychological methods and consideration of the impact of PTSD.

Type
Research Article
Creative Commons
This is a work of the US Government and is not subject to copyright protection within the United States. Published by Cambridge University Press on behalf of International Neuropsychological Society.
Copyright
© U.S. Department of Veterans Affairs, 2024

Introduction

Mild cognitive impairment (MCI) is a condition where individuals exhibit a decline in cognitive functioning greater than that of typical aging but remain independent in their activities of daily living (Petersen et al., Reference Petersen, Lopez, Armstrong, Getchius, Ganguli, Gloss, Gronseth, Marson, Pringsheim, Day, Sager, Stevens and Rae-Grant2018). MCI is often characterized as the early, or prodromal, stage of dementia due to Alzheimer’s disease (AD) or other neurodegenerative or cerebrovascular disease processes (Albert et al., Reference Albert, DeKosky, Dickson, Dubois, Feldman, Fox, Gamst, Holtzman, Jagust, Petersen, Snyder, Carrillo, Thies and Phelps2011). Accurate diagnosis of MCI is critical for early detection and intervention for those at risk for progressive cognitive decline. However, given several diagnostic approaches in use to define MCI, there is unfortunately high variability in its detection across research and clinic samples (Jak et al., Reference Jak, Bondi, Delano-Wood, Wierenga, Corey-Bloom, Salmon and Delis2009; Petersen et al., Reference Petersen, Lopez, Armstrong, Getchius, Ganguli, Gloss, Gronseth, Marson, Pringsheim, Day, Sager, Stevens and Rae-Grant2018).

MCI has typically been defined by the Petersen/Winblad criteria (henceforth referred to as “typical” criteria), which state that an individual has MCI if they are neither cognitively normal nor demented, exhibit cognitive decline from a subjective and objective standpoint, and maintain relatively preserved activities of daily living (Petersen & Morris, Reference Petersen and Morris2005; Petersen, Reference Petersen2004; Winblad et al., Reference Winblad, Palmer, Kivipelto, Jelic, Fratiglioni, Wahlund, Nordberg, Bäckman, Albert, Almkvist, Arai, Basun, Blennow, De Leon, DeCarli, Erkinjuntti, Giacobini, Graff, Hardy, Jack, Jorm, Ritchie, Van Duijn, Visser and Petersen2004). Within this framework, objective cognitive impairment is commonly operationalized as > 1.5 standard deviations (SD) below normative means on at least one neuropsychological measure. The Alzheimer’s Disease Neuroimaging Initiative (ADNI), a large consortium study aimed at validating AD biomarkers and improving treatment trials, uses similar criteria for MCI: subjective memory concerns, impaired score on a paragraph recall test, and intact activities of daily living (Petersen et al., Reference Petersen, Aisen, Beckett, Donohue, Gamst, Harvey, Jack, Jagust, Shaw, Toga, Trojanowski and Weiner2010).

Although efficient, the typical and ADNI criteria for MCI have been critiqued for their tendencies towards misdiagnosis and limited utility in characterizing MCI by type and severity (Bondi et al., Reference Bondi, Edmonds, Jak, Clark, Delano-Wood, McDonald, Nation, Libon, Au, Galasko and Salmon2014; Edmonds et al., Reference Edmonds, Delano-Wood, Galasko, Salmon, Bondi and Initiative2014; Reference Edmonds, Eppig, Bondi, Leyden, Goodwin, Delano-Wood and McDonald2016). Cluster analyses of neuropsychological test scores from individuals in ADNI’s MCI cohort revealed a “cluster-derived normal” subgroup that closely resembled the cognitively unimpaired comparison group in terms of neuropsychological test performance over time, biomarker profiles, and structural neuroimaging (Edmonds et al., Reference Edmonds, Delano-Wood, Galasko, Salmon, Bondi and Initiative2014; Reference Edmonds, Eppig, Bondi, Leyden, Goodwin, Delano-Wood and McDonald2016; Reference Edmonds, Weigand, Thomas, Eppig, Delano-Wood, Galasko, Salmon and Bondi2018). Similarly, a cluster analysis of test scores from individuals who met typical criteria for MCI in a community-based study also revealed a cluster-derived normal group that did not differ from the standardization group on any neuropsychological measures (Clark et al., Reference Clark, Delano-Wood, Libon, McDonald, Nation, Bangen, Jak, Au, Salmon and Bondi2013). These studies suggest that reliance on subjective report of cognitive concerns and a single objective test measure can lead to both false-positive and false-negative errors as well as limitations in standardization, contributing to inaccurate characterization of cognitive status and misdiagnosis.

In light of the high variability in MCI classification, an actuarial method was developed using neuropsychological test data to classify MCI (Bondi et al., Reference Bondi, Jak, Delano-Wood, Jacobson, Delis and Salmon2008; Jak et al., Reference Jak, Bondi, Delano-Wood, Wierenga, Corey-Bloom, Salmon and Delis2009). These neuropsychological criteria for MCI require an individual to score > 1 SD below age-appropriate norms on two tests within a single cognitive domain, or > 1 SD below age-appropriate norms on one test across at least three cognitive domains (Bondi et al., Reference Bondi, Edmonds, Jak, Clark, Delano-Wood, McDonald, Nation, Libon, Au, Galasko and Salmon2014; Jak et al., Reference Jak, Bondi, Delano-Wood, Wierenga, Corey-Bloom, Salmon and Delis2009). The use of multiple tests within and across domains to determine diagnosis was designed to balance sensitivity and reliability. Studies comparing different sets of criteria for MCI found that the neuropsychological criteria yielded more dissociable cognitive phenotypes, significant AD biomarker associations, and stable diagnoses compared to the typical or ADNI criteria (Bondi et al., Reference Bondi, Edmonds, Jak, Clark, Delano-Wood, McDonald, Nation, Libon, Au, Galasko and Salmon2014; Jak et al., Reference Jak, Bondi, Delano-Wood, Wierenga, Corey-Bloom, Salmon and Delis2009). The neuropsychological criteria also better predicted participants’ progression to AD dementia (Bondi et al., Reference Bondi, Edmonds, Jak, Clark, Delano-Wood, McDonald, Nation, Libon, Au, Galasko and Salmon2014). This research highlighted the benefit of applying more comprehensive neuropsychological methods to diagnostic decision-making and the characterization of MCI. Further, it emphasized concerns that prior MCI studies may have underestimated important biomarker relationships through misclassification of participants. A secondary analysis of the Alzheimer’s Disease Cooperative Study (ADCS) donepezil trial found that removal of the cluster-derived normal participants (i.e., false-positive MCI) unmasked beneficial effects of donepezil in terms of lowering the rate of progression to AD (Edmonds et al., Reference Edmonds, Ard, Edland, Galasko, Salmon and Bondi2017).

Biomarker studies, primarily through ADNI, have investigated the utility of cerebrospinal fluid (CSF) and neuroimaging measures for the early identification of AD and evaluation of clinical trials (Ebenau et al., Reference Ebenau, Pelkmans, Verberk, Verfaillie, van den Bosch, van Leeuwenstijn, Collij, Scheltens, Prins, Barkhof, van Berckel, Teunissen and van der Flier2022; Elman et al., Reference Elman, Panizzon, Gustavson, Franz, Sanderson-Cimino, Lyons and Kremen2020; Hansson et al., Reference Hansson, Seibyl, Stomrud, Zetterberg, Trojanowski, Bittner, Lifke, Corradini, Eichenlaub, Batrla, Buck, Zink, Rabe, Blennow and Shaw2018; Veitch et al., Reference Veitch, Weiner, Aisen, Beckett, Cairns, Green, Harvey, Jack, Jagust, Morris, Petersen, Saykin, Shaw, Toga and Trojanowski2019, Reference Veitch, Weiner, Aisen, Beckett, DeCarli, Green, Harvey, Jack, Jagust, Landau, Morris, Okonkwo, Perrin, Petersen, Rivera‐Mindt, Saykin, Shaw, Toga, Tosun and Trojanowski2022). Updated criteria for the AD continuum, including MCI and preclinical AD, have integrated these biomarkers for the detection and prediction of clinical outcomes (Albert et al., Reference Albert, DeKosky, Dickson, Dubois, Feldman, Fox, Gamst, Holtzman, Jagust, Petersen, Snyder, Carrillo, Thies and Phelps2011; McKhann et al., Reference McKhann, Knopman, Chertkow, Hyman, Jack, Kawas, Klunk, Koroshetz, Manly, Mayeux, Mohs, Morris, Rossor, Scheltens, Carrillo, Thies, Weintraub and Phelps2011; Sperling et al., Reference Sperling, Aisen, Beckett, Bennett, Craft, Fagan, Iwatsubo, Jack, Kaye, Montine, Park, Reiman, Rowe, Siemers, Stern, Yaffe, Carrillo, Thies, Morrison‐Bogorad, Wagster and Phelps2011). In 2018, the National Institute on Aging - Alzheimer’s Association put forth a biomarker-based research framework in which the AD continuum was defined using an “ATN” classification system based on the presence of β amyloid deposition (A), phosphorylated tau (T), and neurodegeneration (N) (Jack et al., Reference Jack, Bennett, Blennow, Carrillo, Dunn, Haeberlein, Holtzman, Jagust, Jessen, Karlawish, Liu, Molinuevo, Montine, Phelps, Rankin, Rowe, Scheltens, Siemers, Snyder, Sperling, Masliah, Ryan and Silverberg2018). Biomarkers such as CSF β-amyloid 1–42 (Aβ 42), CSF hyper-phosphorylated tau (p-tau), and CSF total tau (t-tau) can appear abnormal several years before the formal diagnosis of dementia and predict progression to AD dementia (Olsson et al., Reference Olsson, Lautner, Andreasson, Öhrfelt, Portelius, Bjerke, Hölttä, Rosén, Olsson, Strobel, Wu, Dakin, Petzold, Blennow and Zetterberg2016; Shaw et al., Reference Shaw, Vanderstichele, Knapik‐Czajka, Clark, Aisen, Petersen, Blennow, Soares, Simon, Lewczuk, Dean, Siemers, Potter, Lee and Trojanowski2009). However, critiques of a biologically-defined AD state that biomarkers alone are not sufficient to define an individual’s position on the AD continuum without clinical input (Dubois et al., Reference Dubois, Villain, Frisoni, Rabinovici, Sabbagh, Cappa, Bejanin, Bombois, Epelbaum, Teichmann, Habert, Nordberg, Blennow, Galasko, Stern, Rowe, Salloway, Schneider, Cummings and Feldman2021). Rather, in clinical settings, diagnosis should consider the presenting clinical phenotype (e.g., an amnestic syndrome), then examine biomarker positivity.

To date, the majority of research studies examining different MCI criteria and phenotypes in conjunction with AD biomarkers have done so in relatively healthy samples with few comorbidities (Bondi et al., Reference Bondi, Edmonds, Jak, Clark, Delano-Wood, McDonald, Nation, Libon, Au, Galasko and Salmon2014; Edmonds et al., Reference Edmonds, Delano‐Wood, Clark, Jak, Nation, McDonald, Libon, Au, Galasko, Salmon and Bondi2015, Reference Edmonds, Delano-Wood, Galasko, Salmon, Bondi and Initiative2014; Reference Edmonds, Eppig, Bondi, Leyden, Goodwin, Delano-Wood and McDonald2016; Reference Edmonds, Smirnov, Thomas, Graves, Bangen, Delano-Wood, Galasko, Salmon and Bondi2021; Eppig et al., Reference Eppig, Edmonds, Campbell, Sanderson-Cimino, Delano-Wood and Bondi2017; Pommy et al., Reference Pommy, Conant, Butts, Nencka, Wang, Franczak and Glass-Umfleet2023). ADNI, for example, specifically recruited older adults without significant neurological history, psychiatric distress, or high vascular risk (Petersen et al., Reference Petersen, Aisen, Beckett, Donohue, Gamst, Harvey, Jack, Jagust, Shaw, Toga, Trojanowski and Weiner2010). Veteran populations, on the other hand, are typically medically and psychiatrically complicated (e.g., have a high prevalence of post-traumatic stress disorder [PTSD] and history of traumatic brain injury [TBI]) (Carlson et al., Reference Carlson, Kehle, Meis, Greer, MacDonald, Rutks, Sayer, Dobscha and Wilt2011; Greer et al., Reference Greer, Sayer, Spoont, Taylor, Ackland, MacDonald, McKenzie, Rosebush and Wilt2020; Loignon et al., Reference Loignon, Ouellet and Belleville2020; Magruder & Yeager, Reference Magruder and Yeager2009). PTSD has been linked to an increased risk of developing dementia including AD, as well as robust deficits in attention, memory, and processing speed (Desmarais et al., Reference Desmarais, Weidman, Wassef, Bruneau, Friedland, Bajsarowicz, Thibodeau, Herrmann and Nguyen2020; Günak et al., Reference Günak, Billings, Carratu, Marchant, Favarato and Orgeta2020; Scott et al., Reference Scott, Matt, Wrocklage, Crnich, Jordan, Southwick, Krystal and Schweinsburg2015). Similarly, history of TBI has been linked to an increased risk of developing dementia including AD, particularly in Veterans (Gardner et al., Reference Gardner, Bahorik, Kornblith, Allen, Plassman and Yaffe2022; Li et al., Reference Li, Li, Li, Zhang, Zhao, Zhu and Tian2017; Snowden et al., Reference Snowden, Hinde, Reid and Christie2020). Even a history of mild TBI without loss of consciousness in Veterans has been shown to increase the risk for developing dementia (Barnes et al., Reference Barnes, Byers, Gardner, Seal, Boscardin and Yaffe2018). Additionally, Vietnam-era Veterans with histories of TBI have shown elevated levels of CSF p-tau and t-tau, which, in turn, were associated with slower processing speed (Clark et al., Reference Clark, Weigand, Bangen, Thomas, Eglit, Bondi and Delano‐Wood2021). These findings suggest that history of TBI may be associated with pathological brain changes leading to increased risk of dementia.

Given the prevalence of TBI and PTSD in the aging Veteran population, a collaborative study between the Department of Defense and ADNI (DOD-ADNI) was launched to investigate these risk factors for AD in Veterans and their associations with brain AD pathology (Weiner et al., Reference Weiner, Veitch, Hayes, Neylan, Grafman, Aisen, Petersen, Jack, Jagust, Trojanowski, Shaw, Saykin, Green, Harvey, Toga, Friedl, Pacifico, Sheline, Yaffe and Mohlenoff2014). We sought to use the DoD-ADNI database to (1) compare neuropsychological, typical, and ADNI criteria for MCI in a Veteran sample with histories of TBI and/or PTSD in terms of their associations with validated AD biomarkers, and (2) evaluate the effects of TBI and PTSD on diagnosis of MCI by each set of criteria. We predicted that, consistent with past research, the neuropsychological criteria for MCI would show stronger associations with CSF Aβ 42, p-tau181, and t-tau than the typical or ADNI criteria. We also predicted that history of TBI (of any severity) and PTSD symptom severity would both be significant risk factors for MCI by all criteria.

Methods

Data were obtained from the publicly available A Study of Brain Aging in Vietnam War Veterans/DOD-ADNI database (adni.loni.usc.edu). The study is directed by principal investigator Dr Michael Weiner of the San Francisco VA Medical Center and University of California, San Francisco. The overarching goal of the DOD-ADNI study is to investigate the associations between a history of TBI and/or current PTSD and brain AD pathology. The main aims and methods, including participant selection and exclusion criteria, are described in detail elsewhere (Weiner et al., Reference Weiner, Harvey, Hayes, Landau, Aisen, Petersen, Tosun, Veitch, Jack, Decarli, Saykin, Grafman and Neylan2017; Reference Weiner, Veitch, Hayes, Neylan, Grafman, Aisen, Petersen, Jack, Jagust, Trojanowski, Shaw, Saykin, Green, Harvey, Toga, Friedl, Pacifico, Sheline, Yaffe and Mohlenoff2014). Up-to-date information can be found at www.adni-info.org. This research was approved by the Committee on Human Research at the University of California at San Francisco, the San Francisco VA Medical Center Research and Development Committee, and the Department of Defense Human Research Protection Office. Written informed consent was obtained for all study participants. This research was performed in accordance with the ethical standards as laid down in the 1964 Declaration of Helsinki and its later amendments or comparable ethical standards.

Participants

Vietnam-era Veterans completed structured clinical interviews including detailed TBI history, self-report assessments, psychodiagnostic assessment, and neuropsychological assessment. 284 Veterans had complete neuropsychological assessment data and 17 participants were excluded from analyses due to missing or inconsistent TBI or PTSD data, resulting in a sample of 267 Veterans. Sociodemographic information is shown in Table 1. The Clinical Dementia Rating (CDR) Dementia Staging Instrument (Morris, Reference Morris1993) and Mini-Mental State Examination (MMSE) (Folstein et al., Reference Folstein, Folstein and McHugh1975) were performed during screening to rule out significant cognitive or functional impairment indicating dementia (i.e., CDR Global ≥ 1, MMSE < 24).

Table 1. Participant characteristics (n = 267)

CAPS = Clinician-Administered Posttraumatic Stress Disorders Scale for Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, SD = standard deviation, TBI = traumatic brain injury.

TBI and PTSD

Detailed TBI history was obtained using a version of the Ohio State University TBI Identification Method-Interview form. Participants self-reported whether they experienced injuries to the head or neck before, during, and after serving in Vietnam. For each instance of head/neck injury, they provided information about the year of injury, whether they were hospitalized, and the presence and duration of any loss of consciousness (LOC), alteration of consciousness (AOC), or post-traumatic amnesia (PTA). The severity of each injury was classified according to the Veterans Affairs/DoD 2021 Clinical Practice Guidelines (VA/DoD, 2021). An injury was classified as mild if the participant experienced LOC < 30 minutes or AOC or PTA ≤ 24 hours, and moderate/severe if LOC ≥ 30 minutes or AOC or PTA > 24 hours. Current PTSD symptom severity was measured using the Clinician-Administered PTSD Scale for DSM-IV (Blake et al., Reference Blake, Weathers, Nagy, Kaloupek, Charney and Keane1998).

Neuropsychological assessment

Memory functioning for the ADNI criteria was assessed using the Wechsler Memory Scale – Revised Logical Memory II subscale (Delayed Paragraph Recall, Paragraph A only). For the neuropsychological and typical criteria, two measures were used for each domain of memory, language, and processing speed/executive functioning, consistent with past literature and tests available in ADNI protocols (Jak et al., Reference Jak, Bondi, Delano-Wood, Wierenga, Corey-Bloom, Salmon and Delis2009). Memory measures included the Rey Auditory Verbal Learning Test (Schmidt, Reference Schmidt1996) delayed recall and recognition. Language measures included animal fluency and the 30-item Boston Naming Test (Kaplan et al., Reference Kaplan, Goodglass and Weintraub1983). Processing speed/executive functioning measures included the Trail Making Test Parts A and B (Reitan, Reference Reitan1956). Raw scores were converted to z-scores using age, sex, and education-adjusted norms (Stricker et al., Reference Stricker, Christianson, Lundt, Alden, Machulda, Fields, Kremers, Jack, Knopman, Mielke and Petersen2021; Weintraub et al., Reference Weintraub, Salmon, Mercaldo, Ferris, Graff-Radford, Chui, Cummings, DeCarli, Foster, Galasko, Peskind, Dietrich, Beekly, Kukull and Morris2009).

Criteria for MCI

The three sets of diagnostic criteria for MCI are defined in Table 2. Subjective memory concerns were assessed from both the participant and their study partner and were coded as yes/no if either reported concerns. For the neuropsychological and typical criteria, MCI was characterized as amnestic-type if the participant scored below cutoff(s) on memory measures, and non-amnestic if the participant scored below cutoff(s) only on measures on language or processing speed/executive functioning.

Table 2. Diagnostic criteria for mild cognitive impairment

CDR = Clinical Dementia Rating, MMSE = Mini-Mental Status Examination, SD = standard deviation, WMS-R = Wechsler Memory Scale Revised Edition.

Cerebrospinal fluid and genetic markers

A subset (n = 134) of participants completed lumbar puncture to collect CSF samples. CSF levels of p-tau181, t-tau, and Aβ 42 were analyzed using the Roche Elecsys fully automated immunoassay platform and reference LC/MSMS methodology (Kang et al., Reference Kang, Korecka, Figurski, Toledo, Blennow, Zetterberg, Waligorska, Brylska, Fields, Shah, Soares, Dean, Vanderstichele, Petersen, Aisen, Saykin, Weiner, Trojanowski and Shaw2015). Outlier values > 3 SD from the mean for each biomarker were omitted from analyses. Apolipoprotein E (APOE) ε4 positivity was determined by the possession of at least one APOE ε4 allele.

Statistical analyses

Agreement between criteria was assessed using Cohen’s kappa coefficient (κ). Linear multiple regressions adjusting for age and education were conducted to assess associations between MCI status and CSF biomarker by criteria. APOE ε4 positivity was not added as a covariate, as it did not differ between MCI and cognitively normal groups by any criteria. Logistic regressions adjusting for age and education were conducted to simultaneously assess the effects of TBI severity (mild or moderate/severe) and PTSD symptom severity, and potential interactions, on MCI classification by each criteria. Post-hoc analyses assessed the effects of TBI severity and PTSD symptom severity on the subjective and objective components of typical and ADNI criteria.

Results

Identification of MCI

47 (18%) Veterans met neuropsychological criteria, 24 (9%) met typical criteria, and 26 (10%) met ADNI criteria for MCI. The majority of Veterans (201 [75%]) were identified as cognitively normal by all three criteria. Agreement between criteria was poor (Fig. 1). Neuropsychological criteria showed the least agreement with ADNI criteria (κ = 0.11) and minimal agreement with typical criteria (κ = 0.35). Unsurprisingly, typical and ADNI criteria showed the greatest agreement (κ = 0.60) given their common basis of operational definitions. Of those who met neuropsychological criteria for MCI, 31 (66%) were characterized as amnestic MCI (18 single-domain, 13 multi-domain), and 16 (34%) as non-amnestic MCI (13 single-domain, 3 multi-domain). Of those who met typical criteria for MCI, 15 (63%) were characterized as amnestic MCI (7 single-domain, 8 multi-domain), and 9 (37%) as non-amnestic MCI (6 single-domain, 3 multi-domain).

Figure 1. Classification of mild cognitive impairment (MCI) in the DOD-ADNI sample (n = 267) by neuropsychological, typical, and ADNI criteria. CN, cognitively normal; ADNI = Alzheimer’s disease neuroimaging initiative.

Associations with AD biomarkers

MCI diagnosis by neuropsychological criteria was significantly associated with higher p-tau181 and t-tau but was not associated with Aβ 42 (Table 3). MCI diagnosis by typical or ADNI criteria for MCI was not associated with p-tau181, t-tau, or Aβ 42. None of the criteria were associated with p-tau181/Aβ 42 or t-tau/Aβ 42 ratios (all p’s > .05). Subjective memory concerns were not associated with p-tau181, t-tau, or Aβ 42 (all p’s > .15).

Table 3. Multiple linear regressions showing associations between mild cognitive impairment diagnosis by each criteria and cerebrospinal fluid p − tau181, t − tau, and Aβ42 levels after adjusting for age and education

Bolded items indicate p < .05.

Effects of TBI and PTSD on MCI diagnosis

History of mild TBI was not associated with diagnosis of MCI by any criteria (Table 4). History of moderate/severe TBI was associated with MCI by typical criteria and ADNI criteria, but not by neuropsychological criteria. Current PTSD symptom severity was associated with MCI by neuropsychological and ADNI criteria, but not typical criteria (Table 4). Interactions between history of TBI and PTSD symptom severity were not significant (all p’s > .25). Post-hoc analyses found that history of mild TBI (B = 1.06, SE = 0.52, p = .04), history of moderate/severe TBI (B = 1.47, SE = 0.49, p = .003), and PTSD symptom severity (B = 0.53, SE = 0.18, p = .004) were each independently associated with subjective memory concerns. PTSD symptom severity (B = 0.36, SE = 0.14, p = .01), but not history of mild TBI (B = 0.16, SE = 0.34, p = .65) or moderate/severe TBI (B = 0.14, SE = 0.33, p = .68), was associated with scoring > 1.5 SD below demographically adjusted norms on any one test (i.e., objective cognitive impairment by the typical criteria). PTSD symptom severity (B = 0.08, SE = 0.13, p = .52), history of mild TBI (B = 0.21, SE = 0.32, p = .51), and history of moderate/severe TBI (B = 0.22, SE = 0.30, p = .46) were not associated with scoring below cutoff on the Logical Memory Story A recall (i.e., objective memory impairment by the ADNI criteria). In Veterans with history of any TBI, neither time since most recent TBI nor total number of TBIs predicted diagnosis of MCI by any criteria (all p’s > .30).

Table 4. Logistic regressions showing the effects of traumatic brain injury (TBI) and posttraumatic stress disorder (PTSD) severity on diagnosis of mild cognitive impairment by each criteria

CAPS = Clinician-Administered Posttraumatic Stress Disorders Scale for Diagnostic and Statistic Manual of Mental Disorders, Fourth Edition.

Bolded items indicate p < .05.

Discussion

We compared neuropsychological, typical, and ADNI criteria for diagnosing MCI in a complex older-adult Veteran sample. Findings showed that identification of MCI was highly variable with evidenced poor agreement between criteria. Neuropsychological criteria classified the largest number of participants as MCI, nearly twice as much as typical or ADNI criteria. Further, neuropsychological criteria, but not typical or ADNI criteria, were significantly associated with AD biomarkers (e.g., higher CSF p-tau181 and t-tau). History of moderate/severe TBI predicted MCI by typical and ADNI criteria. However, history of moderate/severe TBI was only associated with subjective memory concerns and not with objective impairment as defined by the typical or ADNI criteria, suggesting that the relationship between moderate/severe TBI and MCI by the typical or ADNI criteria was driven largely by subjective memory concerns. Mild TBI did not predict MCI by any criteria. PTSD symptom severity predicted MCI by neuropsychological and ADNI criteria.

The variability seen in MCI classifications is consistent with past research in Veterans, which has found that the percentage of individuals classified as MCI can range from roughly 10%–74% depending on the criteria used (Jak et al., Reference Jak, Bondi, Delano-Wood, Wierenga, Corey-Bloom, Salmon and Delis2009). Relatively low percentages were identified in the current Veteran sample because the DoD-ADNI study initially excluded MCI and dementia in their participant recruitment until a later extension of their study aims. Regardless, neuropsychological criteria classified a greater percentage of participants with MCI of both amnestic and non-amnestic types based on objective cognitive data. ADNI criteria focus on both subjective and objective memory functioning to the exclusion of non-amnestic MCI presentations (e.g., dysnomic, dysexecutive). The typical criteria consider cognitive test performances across domains but use a conservative threshold (>1.5 SD below norms). The inclusion of subjective memory concerns in the typical and ADNI criteria likely contributed to their poor agreement with neuropsychological criteria, as subjective cognitive concerns are only weakly associated with objective performance in healthy older adults and those with MCI (Burmester et al., Reference Burmester, Leathem and Merrick2016). Given these approaches, the ADNI criteria are prone to both false-positive diagnostic errors (due to individuals overestimating their cognitive problems) and false-negative diagnostic errors (Edmonds et al., Reference Edmonds, Delano‐Wood, Clark, Jak, Nation, McDonald, Libon, Au, Galasko, Salmon and Bondi2015, Reference Edmonds, Delano-Wood, Galasko, Salmon, Bondi and Initiative2014; Reference Edmonds, Eppig, Bondi, Leyden, Goodwin, Delano-Wood and McDonald2016). The neuropsychological criteria may have reduced false-positives by requiring multiple low scores on objective tests (e.g., lessening base rates of an impaired score in neurologically normal individuals (Brooks et al., Reference Brooks, Iverson and White2007; Palmer et al., Reference Palmer, Boone, Lesser and Wohl1998)). Neuropsychological criteria may have also reduced false-negatives by capturing individuals with cognitive impairment but without subjective concerns (due to limited insight into their cognitive difficulties or use of sufficient compensatory strategies), who likely would have been overlooked by typical and ADNI approaches.

Stronger associations between neuropsychological criteria and CSF biomarkers of p-tau181 and t-tau, compared to other MCI criteria, were consistent with our hypothesis and past research in ADNI (Bondi et al., Reference Bondi, Edmonds, Jak, Clark, Delano-Wood, McDonald, Nation, Libon, Au, Galasko and Salmon2014; Edmonds et al., Reference Edmonds, Eppig, Bondi, Leyden, Goodwin, Delano-Wood and McDonald2016). Pettigrew et al. (Reference Pettigrew, Soldan, Moghekar, Wang, Gross, O’Brien and Albert2015) similarly found that higher CSF p-tau181 and t-tau were associated with poorer episodic memory in cognitively normal older adults, but there was no relationship between Aβ 42 levels and memory. Generally, increased tau burden has been associated with poorer semantic and episodic memory in older adults with and without MCI (Nathan et al., Reference Nathan, Lim, Abbott, Galluzzi, Marizzoni, Babiloni, Albani, Bartres-Faz, Didic, Farotti, Parnetti, Salvadori, Müller, Forloni, Girtler, Hensch, Jovicich, Leeuwis, Marra, Molinuevo, Nobili, Pariente, Payoux, Ranjeva, Rolandi, Rossini, Schönknecht, Soricelli, Tsolaki, Visser, Wiltfang, Richardson, Bordet, Blin and Frisoni2017; Pelgrim et al., Reference Pelgrim, Beran, Twait, Geerlings and Vonk2021; Reijs et al., Reference Reijs, Ramakers, Köhler, Teunissen, Koel-Simmelink, Nathan, Tsolaki, Wahlund, Waldemar, Hausner, Vandenberghe, Johannsen, Blackwell, Vanderstichele, Verhey and Visser2017). In addition, subjective memory concerns, a key component of the typical and ADNI criteria, were not associated with AD biomarkers. These results suggest that the use of objective cognitive measurements is most aligned with the biological definition of AD while demonstrating clinically relevant cognitive impairment.

A strength of our study was the concurrent analysis of the effects of TBI and PTSD on MCI given their frequent prevalence and co-occurrence in Veterans (Carlson et al., Reference Carlson, Kehle, Meis, Greer, MacDonald, Rutks, Sayer, Dobscha and Wilt2011). While several studies have investigated the effects of head injury/TBI as a risk factor for dementia including AD (Gardner et al., Reference Gardner, Bahorik, Kornblith, Allen, Plassman and Yaffe2022; Li et al., Reference Li, Li, Li, Zhang, Zhao, Zhu and Tian2017), few have examined risk for developing MCI. LoBue et al. (Reference LoBue, Denney, Hynan, Rossetti, Lacritz, Hart, Womack, Woon, Cullum and Abisambra2016) found that in the National Alzheimer’s Coordinating Center database, history of TBI with LOC was associated with increased odds of MCI and earlier diagnosis of MCI, though both effects were substantially attenuated by history of depression and demographic factors (e.g., sex, race). Li et al. (Reference Li, Risacher, McAllister and Saykin2016) similarly found that within the ADNI cohort, history of TBI was associated with earlier age of onset of cognitive impairment, as measured by criteria closely resembling ADNI’s MCI criteria. However, they did not examine the impact of psychiatric factors on diagnosis.

We found that history of moderate/severe, but not mild, TBI was associated with MCI by typical and ADNI criteria. Secondary analyses found that history of mild or moderate/severe TBI was related to subjective concerns but not objective impairment as defined by the typical or ADNI criteria, which is consistent with research showing that the majority of military service members who endorsed subjective memory concerns following mild to severe TBI scored within normal ranges on objective memory tests (French et al., Reference French, Lange and Brickell2014). Subjective cognitive concerns following TBI may bias diagnostic decision-making towards MCI even in the absence of objective cognitive impairment.

We also found that PTSD symptom severity predicted MCI by neuropsychological and ADNI criteria. Studies have found relationships between PTSD severity and increased incidence of MCI in World Trade Center Responders, and increased rate of MCI and dementia diagnoses in Veterans (Bhattarai et al., Reference Bhattarai, Oehlert, Multon and Sumerall2019; Clouston et al., Reference Clouston, Diminich, Kotov, Pietrzak, Richards, Spiro, Deri, Carr, Yang, Gandy, Sano, Bromet and Luft2019). PTSD severity has also been shown to play a larger role than mild TBI in the relationship between subjective cognitive concerns and objective cognitive performance (Mattson et al., Reference Mattson, Nelson, Sponheim and Disner2019). Therefore, PTSD is an important and treatable risk factor to consider when assessing Veterans for neurocognitive disorders.

The DOD-ADNI investigators recently examined prevalence of MCI in the same Veteran cohort as the current study (Weiner et al., Reference Weiner, Harvey, Landau, Veitch, Neylan, Grafman, Aisen, Petersen, Jack, Tosun, Shaw, Trojanowski, Saykin, Hayes and De Carli2023). They identified MCI in a larger proportion of participants (51 out of 289; 18%) and concluded that TBI and PTSD both predicted diagnosis of MCI. The difference in findings is likely attributable to their operationalization of MCI, which differed from our study’s use of the ADNI criteria by adding a telephone screening assessment and ultimately relying on clinician judgment. Although clinician judgment could theoretically integrate subjective and objective information with more nuance than following strict criteria, data-driven diagnoses of MCI can still outperform clinician/consensus diagnoses in terms of capturing individuals with abnormal AD biomarkers that are likely to progress to dementia (Edmonds et al., Reference Edmonds, Smirnov, Thomas, Graves, Bangen, Delano-Wood, Galasko, Salmon and Bondi2021). Weiner and colleagues’ operationalization of TBI also differed from our study such that they categorized participants into groups (e.g., control, TBI, PTSD, or TBI & PTSD) rather than analyzing TBI and PTSD dimensionally.

Limitations of the current study include recruitment by the DOD-ADNI that favored cognitively unimpaired adults, which limited our statistical power in comparing MCI and cognitively unimpaired groups. Additionally, given the sample demographics (e.g., predominantly White, older male Veterans), the generalizability of these results is limited and may not extend to younger, racially/ethnically diverse, female, or civilian groups. Furthermore, this study used a coarse measure of subjective memory concerns, and future research should examine more comprehensive, structured measures of subjective cognition that encompass domains other than memory, such as the Everyday Cognition Test (Farias et al., Reference Farias, Mungas, Harvey, Simmons, Reed and DeCarli2011). TBI characteristics were obtained by retrospective self-report and TBI-related biomarkers such as CSF neurofilament light were not available for analysis. Finally, this study used a cross-sectional analysis that cannot speak to causal relationships between TBI, PTSD, cognition, and AD pathology. Future studies should examine the stability of MCI diagnosis (e.g., reversion rates, progression to AD) by each criteria in the DOD-ADNI sample, expand to other biomarkers of neurodegeneration, and assess potential moderating genetic factors such as APOE ε4 carrier status.

In summary, neuropsychological criteria for diagnosis of MCI appear to be a more sensitive and reliable method of diagnosis that is aligned with biological definitions of early AD compared to typical or ADNI criteria. Diagnostic methods for neurocognitive disorders should incorporate comprehensive neuropsychological methods, particularly for populations with complex medical and psychiatric comorbidities.

Data availability statement

Data from the DOD-ADNI study are publicly available under ADNI’s Data Sharing and Publication policy at adni.loni.usc.edu.

Acknowledgements

Data used in preparation of this article were obtained from the Department of Defense Alzheimer’s Disease Neuroimaging Initiative (DOD-ADNI) database (adni.loni.usc.edu). As such, the investigators within the DOD-ADNI contributed to the design and implementation of the study and/or provided data but did not participate in analysis or writing of this report. A complete listing of ADNI investigators can be found at: http://adni.loni.usc.edu/wp-content/uploads/how_to_apply/ADNI_Acknowledgement_List.pdf. We thank all the veterans for their generous participation in this DOD-ADNI study.

Authorship contribution

M. T. Ly contributed to study design, conducted data analysis and interpretation, and drafted the manuscript. J. Adler contributed to data interpretation, drafting, and revisions of the manuscript. A. Ton Loy contributed to drafting and revisions of the manuscript. E. Edmonds contributed to study design, data interpretation, and revisions of the manuscript. M. Bondi contributed to study design, data interpretation, and revisions of the manuscript. L. Delano-Wood contributed to study design, data interpretation, and revisions of the manuscript.

Funding statement

Data collection and sharing for this project was funded by the Alzheimer’s Disease Neuroimaging Initiative (ADNI) (National Institutes of Health Grant U01 AG024904) and DOD-ADNI (Department of Defense award numbers W81XWH-12-2-0012, W81XWH-13-1-0259, and W81XWH-14-1-0462). ADNI is funded by the National Institute on Aging, the National Institute of Biomedical Imaging and Bioengineering, and through generous contributions from the following: AbbVie, Alzheimer’s Association; Alzheimer’s Drug Discovery Foundation; Araclon Biotech; BioClinica, Inc.; Biogen; Bristol-Myers Squibb Company; CereSpir, Inc.; Cogstate; Eisai Inc.; Elan Pharmaceuticals, Inc.; Eli Lilly and Company; EuroImmun; F. Hoffmann-La Roche Ltd and its affiliated company Genentech, Inc.; Fujirebio; GE Healthcare; IXICO Ltd.; Janssen Alzheimer Immunotherapy Research & Development, LLC.; Johnson & Johnson Pharmaceutical Research & Development LLC.; Lumosity; Lundbeck; Merck & Co., Inc.; Meso Scale Diagnostics, LLC.; NeuroRx Research; Neurotrack Technologies; Novartis Pharmaceuticals Corporation; Pfizer Inc.; Piramal Imaging; Servier; Takeda Pharmaceutical Company; and Transition Therapeutics. The Canadian Institutes of Health Research is providing funds to support ADNI clinical sites in Canada. Private sector contributions are facilitated by the Foundation for the National Institutes of Health (www.fnih.org). The grantee organization is the Northern California Institute for Research and Education, and the study is coordinated by the Alzheimer’s Therapeutic Research Institute at the University of Southern California. DOD-ADNI data are disseminated by the Laboratory for Neuro Imaging at the University of California, Los Angeles.

Competing interests

Dr. Bondi receives royalties from Oxford University Press. The remaining authors have no disclosures to report.

References

Albert, M. S., DeKosky, S. T., Dickson, D., Dubois, B., Feldman, H. H., Fox, N. C., Gamst, A., Holtzman, D. M., Jagust, W. J., Petersen, R. C., Snyder, P. J., Carrillo, M. C., Thies, B., & Phelps, C. H. (2011). The diagnosis of mild cognitive impairment due to Alzheimer’s disease: Recommendations from the national institute on aging-Alzheimer’s association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & Dementia, 7(3), 270279. https://doi.org/10.1016/j.jalz.2011.03.008 CrossRefGoogle ScholarPubMed
Barnes, D. E., Byers, A. L., Gardner, R. C., Seal, K. H., Boscardin, W. J., & Yaffe, K. (2018). Association of mild traumatic brain injury with and without loss of consciousness with dementia in US military veterans. JAMA Neurology, 75(9), 1055. https://doi.org/10.1001/jamaneurol.2018.0815 CrossRefGoogle ScholarPubMed
Bhattarai, J“Jackie”, Oehlert, M. E., Multon, K. D., & Sumerall, S. W. (2019). Dementia and cognitive impairment among U.S. Veterans with a history of MDD or PTSD: A retrospective cohort study based on sex and race. Journal of Aging and Health, 31(8), 13981422. https://doi.org/10.1177/0898264318781131 CrossRefGoogle ScholarPubMed
Blake, D. D., Weathers, F. W., Nagy, L. M., Kaloupek, D. G., Charney, D. S., & Keane, T. M. (1998). Clinician-administered PTSD scale for DSM-IV. National Center for Posttraumatic Stress Disorder.Google Scholar
Bondi, M. W., Edmonds, E. C., Jak, A. J., Clark, L. R., Delano-Wood, L., McDonald, C. R., Nation, D. A., Libon, D. J., Au, R., Galasko, D., Salmon, D. P., & for the Alzheimer’s Disease Neuroimaging Initiative (2014). Neuropsychological criteria for mild cognitive impairment improves diagnostic precision, biomarker associations, and progression rates. Journal of Alzheimer’s Disease, 42(1), 275289. https://doi.org/10.3233/jad-140276 CrossRefGoogle ScholarPubMed
Bondi, M. W., Jak, A. J., Delano-Wood, L., Jacobson, M. W., Delis, D. C., & Salmon, D. P. (2008). Neuropsychological contributions to the early identification of Alzheimer’s disease. Neuropsychology Review, 18(1), 7390. https://doi.org/10.1007/s11065-008-9054-1 CrossRefGoogle Scholar
Brooks, B. L., Iverson, G. L., & White, T. (2007). Substantial risk of “Accidental MCI” in healthy older adults: Base rates of low memory scores in neuropsychological assessment. Journal of the International Neuropsychological Society, 13(3), 490500. https://doi.org/10.1017/s1355617707070531 CrossRefGoogle ScholarPubMed
Burmester, B., Leathem, J., & Merrick, P. (2016). Subjective cognitive complaints and objective cognitive function in aging: A systematic review and meta-analysis of recent cross-sectional findings. Neuropsychology Review, 26(4), 376393. https://doi.org/10.1007/s11065-016-9332-2 CrossRefGoogle ScholarPubMed
Carlson, K. F., Kehle, S. M., Meis, L. A., Greer, N., MacDonald, R., Rutks, I., Sayer, N. A., Dobscha, S. K., & Wilt, T. J. (2011). Prevalence, assessment, and treatment of mild traumatic brain injury and posttraumatic stress disorder. Journal of Head Trauma Rehabilitation, 26(2), 103115. https://doi.org/10.1097/htr.0b013e3181e50ef1 CrossRefGoogle ScholarPubMed
Clark, A. L., Weigand, A. J., Bangen, K. J., Thomas, K. R., Eglit, G. M. L., Bondi, M. W., Delano‐Wood, L., & for the Alzheimer’s Disease Neuroimaging Initiative (2021). Higher cerebrospinal fluid tau is associated with history of traumatic brain injury and reduced processing speed in Vietnam-era veterans: A department of defense Alzheimer’s disease neuroimaging initiative (DOD-ADNI) study. Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring, 13(1), e12239. https://doi.org/10.1002/dad2.12239 Google Scholar
Clark, L. R., Delano-Wood, L., Libon, D. J., McDonald, C. R., Nation, D. A., Bangen, K. J., Jak, A. J., Au, R., Salmon, D. P., & Bondi, M. W. (2013). Are empirically-derived subtypes of mild cognitive impairment consistent with conventional subtypes? Journal of the International Neuropsychological Society, 19(6), 635645. https://doi.org/10.1017/s1355617713000313 CrossRefGoogle ScholarPubMed
Clouston, S. A. P., Diminich, E. D., Kotov, R., Pietrzak, R. H., Richards, M., Spiro, A., Deri, Y., Carr, M., Yang, X., Gandy, S., Sano, M., Bromet, E. J., & Luft, B. J. (2019). Incidence of mild cognitive impairment in World Trade Center responders: Long-term consequences of re-experiencing the events on 9/11/2001. Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring, 11(1), 628636. https://doi.org/10.1016/j.dadm.2019.07.006 Google ScholarPubMed
Desmarais, P., Weidman, D., Wassef, Aéanne, Bruneau, M.-A.ée, Friedland, J., Bajsarowicz, P., Thibodeau, M.-P., Herrmann, N., & Nguyen, Q. D. (2020). The interplay between post-traumatic stress disorder and dementia: A systematic review. The American Journal of Geriatric Psychiatry, 28(1), 4860. https://doi.org/10.1016/j.jagp.2019.08.006 CrossRefGoogle ScholarPubMed
Dubois, B., Villain, N., Frisoni, G. B., Rabinovici, G. D., Sabbagh, M., Cappa, S., Bejanin, A., Bombois, S., Epelbaum, S., Teichmann, M., Habert, M.-O., Nordberg, A., Blennow, K., Galasko, D., Stern, Y., Rowe, C. C., Salloway, S., Schneider, L. S., Cummings, J. L., & Feldman, H. H. (2021). Clinical diagnosis of Alzheimer’s disease: Recommendations of the international working group. The Lancet Neurology, 20(6), 484496. https://doi.org/10.1016/s1474-4422(21)00066-1 CrossRefGoogle ScholarPubMed
Ebenau, J. L., Pelkmans, W., Verberk, I. M. W., Verfaillie, S. C. J., van den Bosch, K. A., van Leeuwenstijn, M., Collij, L. E., Scheltens, P., Prins, N. D., Barkhof, F., van Berckel, B. N. M., Teunissen, C. E., & van der Flier, W. M. (2022). Association of CSF, plasma, and imaging markers of neurodegeneration with clinical progression in people with subjective cognitive decline. Neurology, 98(13), e1315e1326. https://doi.org/10.1212/wnl.0000000000200035 CrossRefGoogle ScholarPubMed
Edmonds, E. C., Ard, M. C., Edland, S. D., Galasko, D. R., Salmon, D. P., & Bondi, M. W. (2017). Unmasking the benefits of donepezil via psychometrically precise identification of mild cognitive impairment: A secondary analysis of the ADCS vitamin E and donepezil in MCI study. Alzheimer’s & Dementia : Translational Research & Clinical Interventions, 4(1), 1118. https://doi.org/10.1016/j.trci.2017.11.001 Google ScholarPubMed
Edmonds, E. C., Delano-Wood, L., Galasko, D. R., Salmon, D. P., Bondi, M. W., & Initiative, A. D. N. (2014). Subjective cognitive complaints contribute to misdiagnosis of mild cognitive impairment. Journal of the International Neuropsychological Society, 20(8), 836847. https://doi.org/10.1017/s135561771400068x CrossRefGoogle ScholarPubMed
Edmonds, E. C., Delano‐Wood, L., Clark, L. R., Jak, A. J., Nation, D. A., McDonald, C. R., Libon, D. J., Au, R., Galasko, D., Salmon, D. P., Bondi, M. W., & Alzheimer’s Disease Neuroimaging Initiative (2015). Susceptibility of the conventional criteria for mild cognitive impairment to false-positive diagnostic errors. Alzheimer’s & Dementia, 11(4), 415424. https://doi.org/10.1016/j.jalz.2014.03.005 CrossRefGoogle ScholarPubMed
Edmonds, E. C., Eppig, J., Bondi, M. W., Leyden, K. M., Goodwin, B., Delano-Wood, L., McDonald, C. R., & For the Alzheimer’s Disease Neuroimaging Initiative (2016). Heterogeneous cortical atrophy patterns in MCI not captured by conventional diagnostic criteria. Neurology, 87(20), 21082116. https://doi.org/10.1212/wnl.0000000000003326 CrossRefGoogle Scholar
Edmonds, E. C., Smirnov, D. S., Thomas, K. R., Graves, L. V., Bangen, K. J., Delano-Wood, L., Galasko, D. R., Salmon, D. P., & Bondi, M. W. (2021). Data-driven vs consensus diagnosis of MCI. Neurology, 97(13), e1288e1299. https://doi.org/10.1212/wnl.0000000000012600 CrossRefGoogle ScholarPubMed
Edmonds, E. C., Weigand, A. J., Thomas, K. R., Eppig, J., Delano-Wood, L., Galasko, D. R., Salmon, D. P.,& Bondi, M. W. (2018). Increasing inaccuracy of self-reported subjective cognitive complaints Over 24 Months in empirically derived subtypes of mild cognitive impairment. Journal of the International Neuropsychological Society, 24(8), 842853. https://doi.org/10.1017/s1355617718000486 CrossRefGoogle ScholarPubMed
Elman, J. A., Panizzon, M. S., Gustavson, D. E., Franz, C. E., Sanderson-Cimino, M. E., Lyons, M. J., & Kremen, W. S. (2020). Amyloid-β positivity predicts cognitive decline but cognition predicts progression to amyloid-β positivity. Biological Psychiatry, 87(9), 819828. https://doi.org/10.1016/j.biopsych.2019.12.021 CrossRefGoogle ScholarPubMed
Eppig, J. S., Edmonds, E. C., Campbell, L., Sanderson-Cimino, M., Delano-Wood, L., Bondi, M. W., & for the Alzheimer’s Disease Neuroimaging Initiative (2017). Statistically derived subtypes and associations with cerebrospinal fluid and genetic biomarkers in mild cognitive impairment: A latent profile analysis. Journal of the International Neuropsychological Society, 23(7), 564576. https://doi.org/10.1017/s135561771700039x CrossRefGoogle ScholarPubMed
Farias, S. T., Mungas, D., Harvey, D. J., Simmons, A., Reed, B. R., & DeCarli, C. (2011). The measurement of everyday cognition: Development and validation of a short form of the everyday cognition scales. Alzheimer’s & Dementia, 7(6), 593601. https://doi.org/10.1016/j.jalz.2011.02.007 CrossRefGoogle Scholar
Folstein, M. F., Folstein, S. E., & McHugh, P. R. (1975). “Mini-mental state”: A practical method for grading the cognitive state of patients for the clinician. Journal of Psychiatric Research, 12(3), 189198. https://doi.org/10.1016/0022-3956(75)90026-6 CrossRefGoogle ScholarPubMed
French, L. M., Lange, R. T., & Brickell, T. (2014). Subjective cognitive complaints and neuropsychological test performance following military-related traumatic brain injury. Journal of Rehabilitation Research and Development, 51(6), 933950. https://doi.org/10.1682/jrrd.2013.10.0226 CrossRefGoogle ScholarPubMed
Gardner, R. C., Bahorik, A., Kornblith, E. S., Allen, I. E., Plassman, B. L., & Yaffe, K. (2022). Systematic review, meta-analysis, and population attributable risk of dementia associated with traumatic brain injury in civilians and veterans. Journal of Neurotrauma, 40(7-8), 620634. https://doi.org/10.1089/neu.2022.0041 CrossRefGoogle ScholarPubMed
Greer, N., Sayer, N. A., Spoont, M., Taylor, B. C., Ackland, P. E., MacDonald, R., McKenzie, L., Rosebush, C., & Wilt, T. J. (2020). Prevalence and severity of psychiatric disorders and suicidal behavior in service members and veterans with and without traumatic brain injury. Systematic Review. Journal of Head Trauma Rehabilitation, 35(1), 113. https://doi.org/10.1097/htr.0000000000000478 CrossRefGoogle ScholarPubMed
Günak, M. M., Billings, J., Carratu, E., Marchant, N. L., Favarato, G., & Orgeta, V. (2020). Post-traumatic stress disorder as a risk factor for dementia: Systematic review and meta-analysis. The British Journal of Psychiatry, 217(5), 600608. https://doi.org/10.1192/bjp.2020.150 CrossRefGoogle ScholarPubMed
Hansson, O., Seibyl, J., Stomrud, E., Zetterberg, H., Trojanowski, J. Q., Bittner, T., Lifke, V., Corradini, V., Eichenlaub, U., Batrla, R., Buck, K., Zink, K., Rabe, C., Blennow, K., Shaw, L. M., & for the Swedish BioFINDER study groupAlzheimer’s Disease Neuroimaging Initiative (2018). CSF biomarkers of Alzheimer’s disease concord with amyloid-β PET and predict clinical progression: A study of fully automated immunoassays in bioFINDER and ADNI cohorts. Alzheimer’s & Dementia, 14(11), 14701481. https://doi.org/10.1016/j.jalz.2018.01.010 CrossRefGoogle ScholarPubMed
Jack, C. R. Jr., Bennett, D. A., Blennow, K., Carrillo, M. C., Dunn, B., Haeberlein, S. B., Holtzman, D. M., Jagust, W., Jessen, F., Karlawish, J., Liu, E., Molinuevo, J. L., Montine, T., Phelps, C., Rankin, K. P., Rowe, C. C., Scheltens, P., Siemers, E., Snyder, H. M., Sperling, R., Masliah, E., Ryan, L., & Silverberg, N. (2018). NIA-AA research framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s & Dementia, 14(4), 535562. https://doi.org/10.1016/j.jalz.2018.02.018 CrossRefGoogle Scholar
Jak, A. J., Bondi, M. W., Delano-Wood, L., Wierenga, C., Corey-Bloom, J., Salmon, D. P., & Delis, D. C. (2009). Quantification of five neuropsychological approaches to defining mild cognitive impairment. The American Journal of Geriatric Psychiatry, 17(5), 368375. https://doi.org/10.1097/jgp.0b013e31819431d5 CrossRefGoogle ScholarPubMed
Kang, J‐Hee, Korecka, M., Figurski, M. J., Toledo, J. B., Blennow, K., Zetterberg, H., Waligorska, T., Brylska, M., Fields, L., Shah, N., Soares, H., Dean, R. A., Vanderstichele, H., Petersen, R. C., Aisen, P. S., Saykin, A. J., Weiner, M. W., Trojanowski, J. Q., Shaw, L. M., & Alzheimer’s Disease Neuroimaging Initiative (2015). The Alzheimer’s disease neuroimaging initiative 2 biomarker core: A review of progress and plans. Alzheimer’s & Dementia, 11(7), 772791. https://doi.org/10.1016/j.jalz.2015.05.003 CrossRefGoogle Scholar
Kaplan, E., Goodglass, H., & Weintraub, S. (1983). Boston Naming Test. APA PsycTests. https://doi.org/10.1037/t27208-000 Google Scholar
Li, W., Risacher, S. L., McAllister, T. W., & Saykin, A. J. (2016). Traumatic brain injury and age at onset of cognitive impairment in older adults. Journal of Neurology, 263(7), 12801285. https://doi.org/10.1007/s00415-016-8093-4 CrossRefGoogle ScholarPubMed
Li, Y., Li, Y., Li, X., Zhang, S., Zhao, J., Zhu, X., & Tian, G. (2017). Head injury as a risk factor for dementia and Alzheimer’s disease: A systematic review and meta-analysis of 32 Observational studies. PLoS ONE, 12(1), e0169650. https://doi.org/10.1371/journal.pone.0169650 CrossRefGoogle ScholarPubMed
LoBue, C., Denney, D., Hynan, L. S., Rossetti, H. C., Lacritz, L. H., Hart, J., Womack, K. B., Woon, F. L., Cullum, C. M., & Abisambra, J. (2016). Self-reported traumatic brain injury and mild cognitive impairment: Increased risk and earlier age of diagnosis. Journal of Alzheimer’s Disease, 51(3), 727736. https://doi.org/10.3233/jad-150895 CrossRefGoogle ScholarPubMed
Loignon, A., Ouellet, M.-C., & Belleville, G. (2020). A systematic review and meta-analysis on PTSD following TBI among military/Veteran and civilian populations. Journal of Head Trauma Rehabilitation, 35(1), E21E35. https://doi.org/10.1097/htr.0000000000000514 CrossRefGoogle ScholarPubMed
Magruder, K. M., & Yeager, D. E. (2009). The prevalence of PTSD across war eras and the effect of deployment on PTSD: A systematic review and meta-analysis. Psychiatric Annals, 39(8), 778788. https://doi.org/10.3928/00485713-20090728-04 CrossRefGoogle Scholar
Mattson, E. K., Nelson, N. W., Sponheim, S. R., & Disner, S. G. (2019). The impact of PTSD and mTBI on the relationship between subjective and objective cognitive deficits in combat-exposed veterans. Neuropsychology, 33(7), 913921. https://doi.org/10.1037/neu0000560 CrossRefGoogle ScholarPubMed
McKhann, G. M., Knopman, D. S., Chertkow, H., Hyman, B. T., Jack, C. R. Jr., Kawas, C. H., Klunk, W. E., Koroshetz, W. J., Manly, J. J., Mayeux, R., Mohs, R. C., Morris, J. C., Rossor, M. N., Scheltens, P., Carrillo, M. C., Thies, B., Weintraub, S., & Phelps, C. H. (2011). The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the national institute on aging-Alzheimer’s association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & Dementia, 7(3), 263269. https://doi.org/10.1016/j.jalz.2011.03.005 CrossRefGoogle ScholarPubMed
Morris, J. C. (1993). The clinical dementia rating (CDR): Current version and scoring rules. Neurology, 43(11), 24122414. https://doi.org/10.1212/wnl.43.11.2412-a CrossRefGoogle ScholarPubMed
Nathan, P. J., Lim, Y. Y., Abbott, R., Galluzzi, S., Marizzoni, M., Babiloni, C., Albani, D., Bartres-Faz, D., Didic, M., Farotti, L., Parnetti, L., Salvadori, N., Müller, B. W., Forloni, G., Girtler, N., Hensch, T., Jovicich, J., Leeuwis, A., Marra, C., Molinuevo, Jé L., Nobili, F., Pariente, J., Payoux, P., Ranjeva, J.-P., Rolandi, E., Rossini, P. M., Schönknecht, P., Soricelli, A., Tsolaki, M., Visser, P. J., Wiltfang, J., Richardson, J. C., Bordet, Régis, Blin, O., & Frisoni, G. B. (2017). Association between CSF biomarkers, hippocampal volume and cognitive function in patients with amnestic mild cognitive impairment (MCI). Neurobiology of Aging, 53, 110. https://doi.org/10.1016/j.neurobiolaging.2017.01.013 CrossRefGoogle ScholarPubMed
Olsson, B., Lautner, R., Andreasson, U., Öhrfelt, A., Portelius, E., Bjerke, M., Hölttä, M., Rosén, C., Olsson, C., Strobel, G., Wu, E., Dakin, K., Petzold, M., Blennow, K., & Zetterberg, H. (2016). CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: A systematic review and meta-analysis. The Lancet Neurology, 15(7), 673684. https://doi.org/10.1016/s1474-4422(16)00070-3 CrossRefGoogle ScholarPubMed
Palmer, B. W., Boone, K. B., Lesser, I. M., & Wohl, M. A. (1998). Base rates of “Impaired” neuropsychological test performance among healthy older adults. Archives of Clinical Neuropsychology, 13(6), 503511. https://doi.org/10.1093/arclin/13.6.503 Google ScholarPubMed
Pelgrim, T. A. D., Beran, M., Twait, E. L., Geerlings, M. I., & Vonk, J. M. J. (2021). Cross-sectional associations of tau protein biomarkers with semantic and episodic memory in older adults without dementia: A systematic review and meta-analysis. Ageing Research Reviews, 71, 101449. https://doi.org/10.1016/j.arr.2021.101449 CrossRefGoogle ScholarPubMed
Petersen, R. C. (2004). Mild cognitive impairment as a diagnostic entity. Journal of Internal Medicine, 256(3), 183194. https://doi.org/10.1111/j.1365-2796.2004.01388.x CrossRefGoogle ScholarPubMed
Petersen, R. C., Aisen, P. S., Beckett, L. A., Donohue, M. C., Gamst, A. C., Harvey, D. J., Jack, C. R. Jr, Jagust, W. J., Shaw, L. M., Toga, A. W., Trojanowski, J. Q., & Weiner, M. W. (2010). Alzheimer’s disease neuroimaging initiative (ADNI) clinical characterization. Neurology, 74(3), 201209. https://doi.org/10.1212/wnl.0b013e3181cb3e25 CrossRefGoogle Scholar
Petersen, R. C., Lopez, O., Armstrong, M. J., Getchius, T. S. D., Ganguli, M., Gloss, D., Gronseth, G. S., Marson, D., Pringsheim, T., Day, G. S., Sager, M., Stevens, J., & Rae-Grant, A. (2018). Practice guideline update summary. Neurology, 90(3), 126135. https://doi.org/10.1212/wnl.0000000000004826 CrossRefGoogle ScholarPubMed
Petersen, R. C., & Morris, J. C. (2005). Mild cognitive impairment as a clinical entity and treatment target. Archives of Neurology, 62(7), 11601163. https://doi.org/10.1001/archneur.62.7.1160 CrossRefGoogle ScholarPubMed
Pettigrew, C., Soldan, A., Moghekar, A., Wang, M.-C., Gross, A. L., O’Brien, R., & Albert, M. (2015). Relationship between cerebrospinal fluid biomarkers of Alzheimer’s disease and cognition in cognitively normal older adults. Neuropsychologia, 78, 6372. https://doi.org/10.1016/j.neuropsychologia.2015.09.024 CrossRefGoogle ScholarPubMed
Pommy, J., Conant, L., Butts, A. M., Nencka, A., Wang, Y., Franczak, M., & Glass-Umfleet, L. (2023). A graph theoretic approach to neurodegeneration: five data-driven neuropsychological subtypes in mild cognitive impairment. In Aging, neuropsychology, and cognition (pp. 120). https://doi.org/10.1080/13825585.2022.2163973 Google Scholar
Reijs, B. L. R., Ramakers, I. H. G. B., Köhler, S., Teunissen, C. E., Koel-Simmelink, M., Nathan, P. J., Tsolaki, M., Wahlund, L.-O., Waldemar, G., Hausner, L., Vandenberghe, R., Johannsen, P., Blackwell, A., Vanderstichele, H., Verhey, F., & Visser, P. J. (2017). Memory correlates of Alzheimer’s disease cerebrospinal fluid markers: A longitudinal cohort study. Journal of Alzheimer’s Disease, 60(3), 11191128. https://doi.org/10.3233/jad-160766 CrossRefGoogle ScholarPubMed
Reitan, R. M. (1956). Trail making test. Manual for administration, scoring, and interpretation: University Press.Google Scholar
Schmidt, M. (1996). Rey auditory verbal learning test: Western Psychological Services.Google Scholar
Scott, J. C., Matt, G. E., Wrocklage, K. M., Crnich, C., Jordan, J., Southwick, S. M., Krystal, J. H., & Schweinsburg, B. C. (2015). A quantitative meta-analysis of neurocognitive functioning in posttraumatic stress disorder. Psychological Bulletin, 141(1), 105140. https://doi.org/10.1037/a0038039 CrossRefGoogle ScholarPubMed
Shaw, L. M., Vanderstichele, H., Knapik‐Czajka, M., Clark, C. M., Aisen, P. S., Petersen, R. C., Blennow, K., Soares, H., Simon, A., Lewczuk, P., Dean, R., Siemers, E., Potter, W., Lee, V. M.‐Y., Trojanowski, J. Q., & Alzheimer’s Disease Neuroimaging Initiative (2009). Cerebrospinal fluid biomarker signature in Alzheimer’s disease neuroimaging initiative subjects. Annals of Neurology, 65(4), 403413. https://doi.org/10.1002/ana.21610 CrossRefGoogle ScholarPubMed
Snowden, T. M., Hinde, A. K., Reid, H. M. O., & Christie, B. R. (2020). Does mild traumatic brain injury increase the risk for dementia? A Systematic Review and Meta-Analysis. Journal of Alzheimer’s Disease, 78(2), 757775. https://doi.org/10.3233/jad-200662 Google ScholarPubMed
Sperling, R. A., Aisen, P. S., Beckett, L. A., Bennett, D. A., Craft, S., Fagan, A. M., Iwatsubo, T., Jack, C. R. Jr., Kaye, J., Montine, T. J., Park, D. C., Reiman, E. M., Rowe, C. C., Siemers, E., Stern, Y., Yaffe, K., Carrillo, M. C., Thies, B., Morrison‐Bogorad, M., Wagster, M. V., & Phelps, C. H. (2011). Toward defining the preclinical stages of Alzheimer’s disease: Recommendations from the national institute on aging-Alzheimer’s association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s & Dementia, 7(3), 280292. https://doi.org/10.1016/j.jalz.2011.03.003 CrossRefGoogle ScholarPubMed
Stricker, N. H., Christianson, T. J., Lundt, E. S., Alden, E. C., Machulda, M. M., Fields, J. A., Kremers, W. K., Jack, C. R. Jr, Knopman, D. S., Mielke, M. M., & Petersen, R. C. (2021). Mayo normative studies: Regression-based normative data for the auditory verbal learning test for ages 30-91 years and the importance of adjusting for sex. Journal of the International Neuropsychological Society, 27(3), 211226. https://doi.org/10.1017/s1355617720000752 CrossRefGoogle ScholarPubMed
VA/DoD Management and Rehabilitation of Post-Acute Mild Traumatic Brain Injury Work Group. (2021). VA/DoD clinical practice guideline for the management and rehabilitation of post-acute mild traumatic brain injury. https://www.healthquality.va.gov/guidelines/Rehab/mtbi/VADoDmTBICPGFinal508.pdf Google Scholar
Veitch, D. P., Weiner, M. W., Aisen, P. S., Beckett, L. A., Cairns, N. J., Green, R. C., Harvey, D., Jack, C. R., Jagust, W., Morris, J. C., Petersen, R. C., Saykin, A. J., Shaw, L. M., Toga, A. W., Trojanowski, J. Q., & Alzheimer’s Disease Neuroimaging Initiative (2019). Understanding disease progression and improving Alzheimer’s disease clinical trials: Recent highlights from the Alzheimer’s disease neuroimaging initiative. Alzheimer’s & Dementia, 15(1), 106152. https://doi.org/10.1016/j.jalz.2018.08.005 CrossRefGoogle ScholarPubMed
Veitch, D. P., Weiner, M. W., Aisen, P. S., Beckett, L. A., DeCarli, C., Green, R. C., Harvey, D., Jack, C. R., Jagust, W., Landau, S. M., Morris, J. C., Okonkwo, O., Perrin, R. J., Petersen, R. C., Rivera‐Mindt, M., Saykin, A. J., Shaw, L. M., Toga, A. W., Tosun, D., Trojanowski, J. Q., & Alzheimer’s Disease Neuroimaging Initiative (2022). Using the Alzheimer’s disease neuroimaging initiative to improve early detection, diagnosis, and treatment of Alzheimer’s disease. Alzheimer’s & Dementia, 18(4), 824857. https://doi.org/10.1002/alz.12422 CrossRefGoogle Scholar
Weiner, M. W., Aisen, P. S., Petersen, R. C. (2020) Alzheimer’s Disease Neuroimaging Initiative 3 (ADNI3) Protocol Version 3.1. https://adni.loni.usc.edu/wp-content/themes/freshnews-dev-v2/documents/consentForms/ADNI3_ProtocolVersion3.1_20201204.pdf Google Scholar
Weiner, M. W., Harvey, D., Hayes, J., Landau, S. M., Aisen, P. S., Petersen, R. C., Tosun, D., Veitch, D. P., Jack, C. R., Decarli, C., Saykin, A. J., Grafman, J., Neylan, T. C., & Department of Defense Alzheimer’s Disease Neuroimaging Initiative (2017). Effects of traumatic brain injury and posttraumatic stress disorder on development of Alzheimer’s disease in Vietnam veterans using the Alzheimer’s disease neuroimaging initiative: Preliminary report. Alzheimer’s & Dementia: Translational Research & Clinical Interventions, 3(2), 177188. https://doi.org/10.1016/j.trci.2017.02.005 Google Scholar
Weiner, M. W., Harvey, D., Landau, S. M., Veitch, D. P., Neylan, T. C., Grafman, J. H., Aisen, P. S., Petersen, R. C., Jack, C. R. Jr, Tosun, D., Shaw, L. M., Trojanowski, J. Q., Saykin, A. J., Hayes, J., De Carli, C., & for the Alzheimer’s Disease Neuroimaging Initiative and the Department of Defense Alzheimer’s Disease Neuroimaging Initiative (2023). Traumatic brain injury and post-traumatic stress disorder are not associated with Alzheimer’s disease pathology measured with biomarkers. Alzheimer’s & Dementia, 19(3), 884895. https://doi.org/10.1002/alz.12712 CrossRefGoogle Scholar
Weiner, M. W., Veitch, D. P., Hayes, J., Neylan, T., Grafman, J., Aisen, P. S., Petersen, R. C., Jack, C., Jagust, W., Trojanowski, J. Q., Shaw, L. M., Saykin, A. J., Green, R. C., Harvey, D., Toga, A. W., Friedl, K. E., Pacifico, A., Sheline, Y., Yaffe, K., Mohlenoff, B., & Department of Defense Alzheimer’s Disease Neuroimaging Initiative (2014). Effects of traumatic brain injury and posttraumatic stress disorder on Alzheimer’s disease in veterans, using the Alzheimer’s disease neuroimaging initiative. Alzheimer’s & Dementia, 10(3), S226S235. https://doi.org/10.1016/j.jalz.2014.04.005 Google ScholarPubMed
Weintraub, S., Salmon, D., Mercaldo, N., Ferris, S., Graff-Radford, N. R., Chui, H., Cummings, J., DeCarli, C., Foster, N. L., Galasko, D., Peskind, E., Dietrich, W., Beekly, D. L., Kukull, W. A., & Morris, J. C. (2009). The Alzheimer’s disease centers’ uniform data set (UDS). Alzheimer Disease & Associated Disorders, 23(2), 91101. https://doi.org/10.1097/wad.0b013e318191c7dd CrossRefGoogle ScholarPubMed
Winblad, B., Palmer, K., Kivipelto, M., Jelic, V., Fratiglioni, L., Wahlund, L‐O., Nordberg, A., Bäckman, L., Albert, M., Almkvist, O., Arai, H., Basun, H., Blennow, K., De Leon, M., DeCarli, C., Erkinjuntti, T., Giacobini, E., Graff, C., Hardy, J., Jack, C., Jorm, A., Ritchie, K., Van Duijn, C., Visser, P., & Petersen, R. C. (2004). Mild cognitive impairment – beyond controversies, towards a consensus: Report of the international working group on mild cognitive impairment. Journal of Internal Medicine, 256(3), 240246. https://doi.org/10.1111/j.1365-2796.2004.01380.x CrossRefGoogle Scholar
Figure 0

Table 1. Participant characteristics (n = 267)

Figure 1

Table 2. Diagnostic criteria for mild cognitive impairment

Figure 2

Figure 1. Classification of mild cognitive impairment (MCI) in the DOD-ADNI sample (n = 267) by neuropsychological, typical, and ADNI criteria. CN, cognitively normal; ADNI = Alzheimer’s disease neuroimaging initiative.

Figure 3

Table 3. Multiple linear regressions showing associations between mild cognitive impairment diagnosis by each criteria and cerebrospinal fluid p − tau181, t − tau, and Aβ42 levels after adjusting for age and education

Figure 4

Table 4. Logistic regressions showing the effects of traumatic brain injury (TBI) and posttraumatic stress disorder (PTSD) severity on diagnosis of mild cognitive impairment by each criteria