Returning Research Results That Indicate Risk of Alzheimer Disease Dementia to Healthy Participants in Longitudinal Studies (WeSHARE) ===================================================================================================================================== * Sarah M. Hartz * Spondita Goswami * Amy Oliver * Alissa Evans * Sacha Jackson * Erin Linnenbringer * John C. Morris * Jessica Mozersky ## Abstract **Introduction** Returning research results that indicate risk of Alzheimer disease (AD)-a disease for which no meaningful treatments or cure exist-to cognitively normal participants is controversial. AD is thought to begin many years before clinical signs and symptoms begin. During this time, individuals are cognitively normal, but have biomarkers that indicate pathophysiological changes in the brain. With this study, we aim to evaluate impact of returning research results on cognitively normal participants recruited from a longitudinal observational cohort on aging at the Knight Alzheimer Research Center (Knight ADRC) at Washington University in St. Louis. **Methods and analysis** Our study uses a 2-year, delayed-start randomized clinical trial design. Participants are randomized to receive their research results either in 2-4 weeks or 1 year after informed consent. We plan to approach approximately 260 participants who have research results from previous genetic and biomarker testing at the Knight ADRC. The primary cognitive outcomes are 1-year change in subjective cognitive score (Clinical Dementia Rating (CDR) ® sum of boxes), objective cognitive score (psychometric composite score), and the primary psychosocial outcome is Impact of Event-Revised (IES_R) score 1 year after return of research results. **Ethics and dissemination** This study has been approved by WUSM Institutional Review Board and the Human Research Protection Office. Results from these trials are shared through conferences and publications. **Trial registration number** NCT04699786. ## INTRODUCTION Alzheimer disease (AD) is a progressive neurodegenerative disorder characterized by a prolonged preclinical phase before significant cognitive or behavioral symptoms emerge. The clinical manifestation of the disorder is characterized by cognitive deterioration, changes in behavior and increased requirement for care due to loss of functional independence. Before these signs appear, the brain undergoes structural and functional changes triggered by biomarkers such as amyloid beta (Aβ) accumulation and neurofibrillary tangle pathology. AD biomarkers can be detected years before any clinical symptoms emerge, known as the preclinical phase.1-3 This preclinical phase, characterized by biomarker evidence of AD neuropathology, provides an opportunity for potential clinical intervention, thereby improving our understanding of the disease. There is considerable interest in learning AD biomarker status among research participants,4-6 although limited data exist related to into the impact of disclosing such research results to cognitively normal participants, especially those enrolled in longitudinal studies where there may not be any immediate actionability of knowing one’s biomarker status such as entering a clinical trial. Current research efforts are focused on identify and characterizing biomarkers that are associated with the onset of AD or an increased risk of developing AD, often utilizing longitudinal cohorts that enable individuals to be followed over time. Such research specifically aims to enroll individuals with normal cognitive function but who exhibit brain pathology or biomarkers that indicate brain pathophysiological changes that increase risk of developing AD.3, 7-12 Ethical considerations arise when disclosing biomarker research results to clinically normal participants due to concerns of potential negative consequences arising from such disclosure. These concerns include psychological harms such as anxiety or depression and spurious cognitive test scores due to subjective feelings of improved or worsening memory arising from the knowledge of personal AD risks. However, there is no definitive evidence suggesting that these negative psychological or cognitive effects are significant or long-lasting.13-18 This study aims to bridge this gap by providing a protocol to disclose research results to participants while measuring the psychological and cognitive effects on cognitively normal individuals enrolled in a longitudinal cohort of aging. In addition, the protocol will also delineate steps to examine the actual uptake of research results, including who declines to learn their research results and the process of qualitatively evaluating the reasons behind participants’ decisions to receive/decline their results. These findings will help in providing a framework for safely, ethically, and respectfully disclosing research results that honor participants’ autonomy. Considering the benefits of sharing research results with participants, it is crucial to investigate its impact on outcomes and understand who opts in and out of RoRR. However, the available evidence concerning psychological and behavioral outcomes and cognitive decline after receiving combination of risk research results and an actual number of participants’ uptake of RoRR is limited. The limiting factors include current studies focusing on short-term effects, methodological differences between studies that hinder comparisons, no data on who opts in or out of RoRR, use of returning single risk results and a need for longitudinal follow-up in real research settings. Studies addressing these limitations would facilitate a clear understanding of how RoRR can impact outcomes. More importantly, there still needs to be more evidence about the optimal way of disclosing estimated risk results of combined biomarkers, should clinical breakthroughs suggest disclosure as a standard policy in future studies. This protocol paper outlines this process involved in RoRR regarding AD risk of developing symptomatology on psychological, behavioral, and cognitive outcomes. Additionally, the paper will explain how the number of participants’ uptake of research results will be measured. ## METHODS AND ANALYSIS The trial’s objective is to identify the most effective approach for RoRR to participants, incorporating personalized 5-year AD risk combining predictions based on *APOE* genotyping, recent imaging (Amyloid PET and MRI), age, sex, and race. The aim is to develop a process that minimizes potential psychological and behavioral outcomes and cognitive decline resulting from learning these results. We will use a delayed start randomized noninferiority clinical trial, determine the impact of returning research results that indicate AD risk on cognitive and psychosocial outcomes. Given the limited literature on informing research participants about their estimated AD symptom risk, the hypothesis is that any significant cognitive or psychosocial changes will normalize within a year. Consequently, individuals in the RoRR arm are expected not to differ substantially from those in the delayed-start arm at the 12-month mark, just before the delayed-start arm receives their results. ### Inclusion and exclusion criteria Participants eligible for inclusion in the study will need to meet several criteria. Firstly, they will need to be current Knight ADRC participants who have been classified as cognitively normal (Clinical Dementia Rating19, 20 (CDR®) = 0) within the past 12 months. Additionally, they will need to be aged 65 years or older. Furthermore, participants will need to fulfill one of two conditions: they will need to have had a brain MRI and PET amyloid scan (ideally within the past two years, but up to five years due to COVID-19-related delays will be acceptable), or they will have had their blood drawn for plasma biomarker measurements during an in-person clinical assessment. Moreover, participants will need to have available genetic research results, including their apoE status, and they will need to currently consent to be contacted for other research opportunities through the Knight ADRC. There will be no specific exclusion criteria other than not meeting the inclusion criteria or if a participant does not consent. ### Participants All Knight ADRC participants in longitudinal studies of aging who fulfill the inclusion criteria will be recruited for the study. Prior to scheduling the annual cognitive visit, eligible participants will be mailed a letter informing them of their eligibility, educational materials, and a consent form to review. Approximately two weeks after the study packets are mailed, a study team member will call each potential participant to ask if they have any additional questions based on the information they received and whether they are interested in participating. If participants agree, they will be scheduled for their first visit. A study coordinator will conduct the first visit over Zoom-HIPAA or in person at the Knight ADRC. During this visit, the study coordinator will go through the educational materials, examples of research results, and their limitations, and will obtain informed consent. The educational materials will provide information on the risk factors associated with AD dementia, including age, family history, gender, and educational attainment. Additionally, other risk factors, such as those identified through research, will be introduced, with a reminder that having these factors does not guarantee developing AD dementia. Participants will be informed about the possibility of receiving increased or not increased risk, explaining what these results signify and discussing their limitations. Emphasis will be placed on the fact that the research risk estimate predicts the likelihood of experiencing early symptoms of AD dementia. The informed consent process will be completed if the participant chooses to move forward, and the study coordinator has no concerns about psychiatric stability. After enrollment, using a 2-year delayed-start randomized clinical trial design, participants will be randomized to intervention or delayed arm groups. Participants who decline to receive results will be invited to participate in an interview discussion about their reasons for opting not to receive their research results. Participants who agree to participate in the interview will be contacted by a research team member for their RORR and interview, during which the participant will receive verbal consent. ### Disclosure process After consent from participants in visit one, the intervention group will be scheduled immediately (later for the delayed arm group) for RoRR in the second visit with a genetic counselor or psychiatrist for disclosure. Participants will learn about three key pieces of information. First, their baseline absolute risk of developing Alzheimer’s disease (AD) will be addressed, considering their demographic characteristics. Then, the disclosure process will shift to the adjusted absolute risk of AD based on the research results, with a direct comparison made between the adjusted result and the baseline result. This will include an explanation that the range of adjusted risk represents the best estimates derived from current data. Following this, a review of the research results contributing to the adjusted risk will be discussed. This will include discussing biomarker test results (such as PET Amyloid and MRI or an Amyloid blood test) and the APOE genetic test result. Finally, there will be room for additional discussion and questions to ensure the participant comprehensively understands and can engage with the information provided. Disclosure sessions will be one hour, allowing time for questions and education, and the length will be tracked. Participants will be called by the nurse one week after receiving results to answer any questions or concerns they may have. ## Data collection Data will be collected through surveys using validated measures and semi-structured interviews with participants. Specifically, semi-structured interviews will be conducted with participants who either received or opted out of receiving research results. Surveys will be employed for quantitative assessments of subjective measures of cognitive and psychological outcomes and objective measure of cognitive outcome at baseline, 2 months, 6 months, 12 months, and 24 months. Participant counts will be utilized to determine the number of participants who opted to either receive or not receive research results. Survey instruments include published quantitative measures (including decision to receive results, objective and subjective cognitive scores, impact of event, AD related distress, decisional regret, depression, feelings about results, healthcare utilization, lifestyle/health behavior change, satisfaction with RORR, personal utility, and comprehension of results) will be administered at the time of enrollment (T1), two-month post disclosure/visit (T2), six-months post disclosure/visit (T6), and 12 months post disclosure/visit (T12) or 24 months for delayed group post disclosure/visit (T24) for the two study groups. Longitudinal evaluation of a subset of these measures will enable exploration of changes over time. Table 1 summarizes the primary outcomes and published measures collected in each study group. To ensure a satisfactory response rate, surveys will be offered via multiple modalities, including phone, internet and mail. View this table: [Table 1:](http://medrxiv.org/content/early/2024/07/03/2024.07.01.24309801/T1) Table 1: Schedule of outcomes and measures in quantitative surveys and constructs in qualitative Interviews ## Data analysis We hypothesize that significant cognitive or psychosocial changes will normalize after one year, leading to no substantive clinical differences between individuals in the RoRR arm and those in the delayed-start arm at the 12-month time point (prior to the delayed-start arm receiving results). We will analyze cognitive or psychosocial changes based on RoRR, stratifying by whether the participant received increased risk results. The primary cognitive outcomes include the 1-year change in subjective cognitive score (CDR sum of boxes) and objective cognitive score (psychometric composite), while the primary psychosocial outcome is the impact of the event score at 1 year. Our primary analyses will involve logistic regressions comparing the RoRR arm to the delayed-start arm at 12 months, stratified by risk (increased risk vs. no change or decreased risk) and adjusted for age, sex, and race. Secondary statistical analyses will use logistic and/or linear regressions to determine the effect of RoRR on the other outcomes (see Table 2). We will also explore changes in participation rates potentially due to RoRR and the impact of RoRR on available study partners/families in terms of decisional regret and feelings about the result. ### Psychological support Given concerns about the potential for adverse psychosocial outcomes of RoRR, study staff administering instruments and scoring quantitative measures and those performing qualitative interviews will notify a board-certified psychiatrist (Dr. Sarah Hartz, CO PI of the study) and a neurologist of any clinically relevant scale scores or psychological concerns that arise during data collection and/or results disclosure. Moreover, a genetic counselor will check in with all participants receiving AD risk results two weeks post-disclosure (Groups 1 and 2), and participants who exhibit clinically significant distress or other psychological outcomes will schedule separate therapeutic interactions with Dr. Hartz. Any unanticipated adverse events will be reported to the IRB, and all adverse events (anticipated or unanticipated, serious or not, related or unrelated) will be reported to the funding agency. Based on prior studies of the return of results, we do not anticipate any new-onset depression or suicidality, although it is important to ensure that this is monitored. ## Discussion Returning AD risk results to cognitively normal participants has become increasingly important due to several factors, including maintaining participant autonomy, improving clinical care, and including high-risk participants in clinical treatment studies. Currently, there is a debate about disclosing AD risk results due to limited evidence regarding psychological and cognitive decline. Therefore, data is needed to inform this discussion and shape policies, protocols, and clinical care. This mixed-methods, longitudinal delayed start noninferiority study aims to address this evidentiary gap. Psychological and cognitive data will allow for comparisons between individuals who receive results immediately or on a delay and those who decline to receive results. This study is the first to return multiple types of research results—genetic and biomarker-based—that indicate the risk of AD. Prior studies have evaluated the impact of returning a single research result in specific contexts, but no studies have evaluated the impact of returning multiple research results synthesized into a single estimate of AD risk, which will be necessary as precision medicine evolves and data volume increases. Additionally, this study will provide evidence on the actual uptake of AD risk research results in a real-world setting, considering the potential gap between intentions and behaviors observed in prior genetic testing contexts. Furthermore, this study will examine the impact of RoRR on family members/study partners, as their reactions and needs are crucial. Understanding the impact of RoRR on cognitive outcomes will inform future study designs. Lastly, we will develop educational materials and a training module for healthcare providers based on existing protocols used in AD settings, adapting and expanding these protocols to include different types of research results. We plan to return research, not clinical, results that meet the quality threshold recommended by the National Academy of Sciences (NAS). NAS guidelines state that laboratory analysis must provide confidence in the result, especially when results are not intended for clinical decision-making, as in our study. Our research uses state-of-the-art imaging and genotyping technology to ensure quality and accuracy. Results will be presented as a single risk estimate representing a 5-year risk of developing symptomatic AD, incorporating published AD incidence rates and risk curves. This flexible approach can accommodate new biomarkers as evidence emerges. ## Dissemination We plan to continue to share the results of this trial through local, national and international conferences and publications. ## Data Availability All data produced in the present study are available upon reasonable request to the authors * Received July 1, 2024. * Revision received July 1, 2024. * Accepted July 3, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.Epelbaum S, Genthon R, Cavedo E, et al. Preclinical Alzheimer’s disease: A systematic review of the cohorts underlying the concept. Alzheimer’s & dementia : the journal of the Alzheimer’s Association. Apr 2017;13(4):454–467. doi:10.1016/j.jalz.2016.12.003 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jalz.2016.12.003&link_type=DOI) 2. 2.Sperling RA, Aisen PS, Beckett LA, et al. 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 : the journal of the Alzheimer’s Association. May 2011;7(3):280–92. doi:10.1016/j.jalz.2011.03.003 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jalz.2011.03.003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21514248&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000291239600005&link_type=ISI) 3. 3.Sperling RA, Karlawish J, Johnson KA. Preclinical Alzheimer disease-the challenges ahead. Nat Rev Neurol. Jan 2013;9(1):54–8. doi:10.1038/nrneurol.2012.241 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrneurol.2012.241&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23183885&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) 4. 4.Botkin JR, Mancher M, Busta ER, Downey AS. Returning individual research results to participants: guidance for a new research paradigm. The National Academies Press; 2018. 5. 5.Walter S, Taylor A, Tyrone J, et al. Disclosing Individual Results in Dementia Research: A Proposed Study Participant’s Bill of Rights. Journal of Alzheimer’s disease : JAD. 2022;90(3):945–952. doi:10.3233/JAD-220810 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3233/JAD-220810&link_type=DOI) 6. 6.Gooblar J, Roe CM, Selsor NJ, Gabel MJ, Morris JC. Attitudes of Research Participants and the General Public Regarding Disclosure of Alzheimer Disease Research Results. JAMA neurology. Dec 2015;72(12):1484–90. doi:10.1001/jamaneurol.2015.2875 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jamaneurol.2015.2875&link_type=DOI) 7. 7.Grill JD, Galvin JE. Facilitating Alzheimer disease research recruitment. Alzheimer Dis Assoc Disord. Jan-Mar 2014;28(1):1–8. doi:10.1097/WAD.0000000000000016 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/WAD.0000000000000016&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24322484&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) 8. 8.Sperling RA, Rentz DM, Johnson KA, et al. The A4 study: stopping AD before symptoms begin? Science translational medicine. Mar 19 2014;6(228):228fs13. doi:10.1126/scitranslmed.3007941 [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjEzOiI2LzIyOC8yMjhmczEzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDcvMDMvMjAyNC4wNy4wMS4yNDMwOTgwMS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 9. 9.GeneMatch: Connecting Alzheimer’s Prevention Studies with Eligible Volunteers. [https://www.endalznow.org/genematch](https://www.endalznow.org/genematch) 10. 10.Alzheimer’s Prevention Registry. [https://www.endalznow.org/](https://www.endalznow.org/) 11. 11.The Dominantly Inherited Alzheimer Network. Washington University School of Medicine St Louis. [https://dian.wustl.edu/our-research/observational-study/](https://dian.wustl.edu/our-research/observational-study/) 12. 12.Aisen PS, Jimenez-Maggiora GA, Rafii MS, Walter S, Raman R. Early-stage Alzheimer disease: getting trial-ready. Nature Reviews Neurology. 2022/07/01 2022;18(7):389–399. doi:10.1038/s41582-022-00645-6 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41582-022-00645-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35379951&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) 13. 13.Paulsen JS, Nance M, Kim JI, et al. A review of quality of life after predictive testing for and earlier identification of neurodegenerative diseases. Prog Neurobiol. Nov 2013;110:2–28. doi:10.1016/j.pneurobio.2013.08.003 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pneurobio.2013.08.003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24036231&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) 14. 14.de Wilde A, van Buchem MM, Otten RHJ, et al. Disclosure of amyloid positron emission tomography results to individuals without dementia: a systematic review. Alzheimers Res Ther. Jul 28 2018;10(1):72. doi:10.1186/s13195-018-0398-3 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13195-018-0398-3&link_type=DOI) 15. 15.Lim YY, Maruff P, Getter C, Snyder PJ. Disclosure of positron emission tomography amyloid imaging results: A preliminary study of safety and tolerability. Alzheimer’s & dementia : the journal of the Alzheimer’s Association. Apr 2016;12(4):454–8. doi:10.1016/j.jalz.2015.09.005 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jalz.2015.09.005&link_type=DOI) 16. 16.Burns JM, Johnson DK, Liebmann EP, Bothwell RJ, Morris JK, Vidoni ED. Safety of disclosing amyloid status in cognitively normal older adults. Alzheimer’s & dementia : the journal of the Alzheimer’s Association. Sep 2017;13(9):1024–1030. doi:10.1016/j.jalz.2017.01.022 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jalz.2017.01.022&link_type=DOI) 17. 17.Green RC, Roberts JS, Cupples LA, et al. Disclosure of APOE genotype for risk of Alzheimer’s disease. N Engl J Med. Jul 16 2009;361(3):245–54. doi:10.1056/NEJMoa0809578 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa0809578&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19605829&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000267976100005&link_type=ISI) 18. 18.Green RC, Christensen KD, Cupples LA, et al. A randomized noninferiority trial of condensed protocols for genetic risk disclosure of Alzheimer’s disease. Alzheimer’s & dementia : the journal of the Alzheimer’s Association. Oct 2015;11(10):1222–30. doi:10.1016/j.jalz.2014.10.014 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jalz.2014.10.014&link_type=DOI) 19. 19.Morris JC. The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology. Nov 1993;43(11):2412–4. doi:10.1212/wnl.43.11.2412-a [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1212/WNL.43.11.2412-a&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8232972&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) 20. 20.Morris JC, Ernesto C, Schafer K, et al. Clinical dementia rating training and reliability in multicenter studies: the Alzheimer’s Disease Cooperative Study experience. Neurology. Jun 1997;48(6):1508–10. doi:10.1212/wnl.48.6.1508 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1212/WNL.48.6.1508&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9191756&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) 21. 21.Ritter PL, Stewart AL, Kaymaz H, Sobel DS, Block DA, Lorig KR. Self-reports of health care utilization compared to provider records. J Clin Epidemiol. Feb 2001;54(2):136–41. doi:10.1016/s0895-4356(00)00261-4 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0895-4356(00)00261-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11166528&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000166819400006&link_type=ISI) 22. 22.Roberts JS, Cupples LA, Relkin NR, Whitehouse PJ, Green RC. Genetic Risk Assessment for Adult Children of People With Alzheimer’s Disease: The Risk Evaluation and Education for Alzheimer’s Disease (REVEAL) Study. Journal of Geriatric Psychiatry and Neurology. 2005/12/01 2005;18(4):250–255. doi:10.1177/0891988705281883 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/0891988705281883&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16306249&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000233433100014&link_type=ISI) 23. 23.Roberts JS, Connell CM. Illness representations among first-degree relatives of people with Alzheimer disease. Alzheimer Dis Assoc Disord. Jul-Sep 2000;14(3):129–136, Discussion 127-8. doi:10.1097/00002093-200007000-00003 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/00002093-200007000-00003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10994653&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000089110600003&link_type=ISI) 24. 24.Li M, Bennette CS, Amendola LM, et al. The Feelings About genomiC Testing Results (FACToR) Questionnaire: Development and Preliminary Validation. Journal of genetic counseling. Sep 1 2018; doi:10.1007/s10897-018-0286-9 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10897-018-0286-9&link_type=DOI) 25. 25.Mazor KM, Street RL, Jr., Sue VM, Williams AE, Rabin BA, Arora NK. Assessing patients’ experiences with communication across the cancer care continuum. Patient education and counseling. Aug 2016;99(8):1343–8. doi:10.1016/j.pec.2016.03.004 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pec.2016.03.004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26979476&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) 26. 26.Street RL, Jr., Mazor KM, Arora NK. Assessing Patient-Centered Communication in Cancer Care: Measures for Surveillance of Communication Outcomes. Journal of oncology practice. Dec 2016;12(12):1198–1202. doi:10.1200/JOP.2016.013334 [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoiam9wIjtzOjU6InJlc2lkIjtzOjE3OiJKT1AuMjAxNi4wMTMzMzR2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzA3LzAzLzIwMjQuMDcuMDEuMjQzMDk4MDEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 27. 27.1. Wilson J, 2. Keane TM Weiss DS, Marmar CR. The Impact of Event Scale - Revised. In: Wilson J, Keane TM, eds. Assessing psychological trauma and PTSD. Guilford; 1996:399–411. 28. 28.Brehaut JC, O’Connor AM, Wood TJ, et al. Validation of a decision regret scale. Medical decision making : an international journal of the Society for Medical Decision Making. Jul-Aug 2003;23(4):281–92. doi:10.1177/0272989X03256005 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/0272989X03256005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12926578&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000184395300002&link_type=ISI) 29. 29.Amendola LM, Berg JS, Horowitz CR, et al. The Clinical Sequencing Evidence-Generating Research Consortium: Integrating Genomic Sequencing in Diverse and Medically Underserved Populations. The American Journal of Human Genetics. 2018;103(3):319–327. doi:10.1016/j.ajhg.2018.08.007 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajhg.2018.08.007&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) 30. 30.Lang FR, Carstensen LL. Time counts: future time perspective, goals, and social relationships. Psychology and aging. Mar 2002;17(1):125–39. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1037/0882-7974.17.1.125&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11931281&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000174439300010&link_type=ISI) 31. 31.Link BG, Struening EL, Rahav M, Phelan JC, Nuttbrock L. On stigma and its consequences: evidence from a longitudinal study of men with dual diagnoses of mental illness and substance abuse. Journal of health and social behavior. Jun 1997;38(2):177–90. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2307/2955424&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9212538&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1997XG85800006&link_type=ISI) 32. 32.Pan A-W, Chung, LyInn, Fife, Betsy, Hsiung, Ping-Chuan. Evaluation of the psychometrics of the Social Impact Scale: a measure of stigmatization. International Journal of Rehabilitation Research. 2007;30((3)):235–238. doi:10.1097 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/MRR.0b013e32829fb3db&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17762770&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) 33. 33.Fife BL, Wright ER. The dimensionality of stigma: a comparison of its impact on the self of persons with HIV/AIDS and cancer. Journal of health and social behavior. Mar 2000;41(1):50–67. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2307/2676360&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10750322&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000086059200004&link_type=ISI) 34. 34.Lesher EL, Berryhill JS. Validation of the Geriatric Depression Scale--Short Form among inpatients. J Clin Psychol. Mar 1994;50(2):256–60. doi:10.1002/1097-4679(199403)50:2<256::aid-jclp2270500218>3.0.co;2-e [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/1097-4679(199403)50:2<256::AID-JCLP2270500218>3.0.CO;2-E&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8014251&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F03%2F2024.07.01.24309801.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1994NE67500017&link_type=ISI)